Language selection

Search

Patent 2930925 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2930925
(54) English Title: BIOMARKER SIGNATURE METHOD, AND APPARATUS AND KITS THEREFOR
(54) French Title: PROCEDE DE SIGNATURE DE BIOMARQUEURS, ET APPAREIL ET KITS ASSOCIES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • G01N 33/50 (2006.01)
  • G06F 19/18 (2011.01)
  • C12Q 1/68 (2006.01)
(72) Inventors :
  • BRANDON, RICHARD BRUCE (Australia)
  • MCHUGH, LEO CHARLES (United States of America)
(73) Owners :
  • IMMUNEXPRESS PTY LTD (Australia)
(71) Applicants :
  • IMMUNEXPRESS PTY LTD (Australia)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2015-02-06
(87) Open to Public Inspection: 2015-08-13
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/AU2015/050043
(87) International Publication Number: WO2015/117204
(85) National Entry: 2016-05-17

(30) Application Priority Data:
Application No. Country/Territory Date
2014900363 Australia 2014-02-06

Abstracts

English Abstract

The present invention discloses methods, kits, and apparatus as well as reagents and compositions associated therewith for deriving an indicator for use in diagnosing the presence, absence or degree of at least one condition in a biological subject or in prognosing at least one condition in a biological subject. Also disclosed is a biomarker signature for use in diagnosing the presence, absence or degree of at least one condition in a biological subject or in prognosing at least one condition in a biological subject. The present invention further discloses methods, kits and apparatus, as well as reagents and compositions associated therewith, for identifying biomarkers for use in a biomarker signature.


French Abstract

La présente invention concerne des procédés, des kits et un appareil, ainsi que des réactifs et des compositions associés à ceux-ci pour élaborer un indicateur destiné à être utilisé lors du diagnostic de la présence, de l'absence ou du degré d'au moins une affection chez un sujet biologique ou lors du pronostic d'au moins une affection chez un sujet biologique. L'invention concerne également une signature de biomarqueurs destinée à être utilisée lors du diagnostic de la présence, de l'absence ou du degré d'au moins une affection chez un sujet biologique ou lors du pronostic d'au moins une affection chez un sujet biologique. La présente invention concerne en outre des procédés, des kits et un appareil, ainsi que des réactifs et des compositions associés à ceux-ci, servant à identifier des biomarqueurs en vue de les utiliser dans une signature de biomarqueurs.

Claims

Note: Claims are shown in the official language in which they were submitted.


- 230 -

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
1. A method for determining an indicator used in assessing a likelihood of a
biological
subject having a presence, absence, degree or prognosis of at least one
medical condition,
the method including:
a) determining a pair of biomarker values, each biomarker value being a value
measured
or derived for at least one corresponding immune system biomarker of the
biological
subject and being at least partially indicative of a concentration of the
immune system
biomarker in a sample taken from the subject;
b) determining a derived biomarker value using the pair of biomarker values,
the derived
biomarker value being indicative of a ratio of concentrations of the pair of
immune
system biomarkers; and,
c) determining the indicator using the derived biomarker value.
2. A method according to claim 1, wherein the method includes:
a) determining a first derived biomarker value using a first pair of biomarker
values, the
first derived biomarker value being indicative of a ratio of concentrations of
first and
second immune system biomarkers;
b) determining a second derived biomarker value using a second pair of
biomarker
values, the second derived biomarker value being indicative of a ratio of
concentrations of third and fourth immune system biomarkers; and,
c) determining the indicator by combining the first and second derived
biomarker
values.
3. A method according to claim 2, wherein the method includes combining the
derived
biomarker values using a combining function, the combining function being at
least one
of:
a) an additive model;
b) a linear model;
c) a support vector machine;
d) a neural network model;
e) a random forest model;
f) a regression model;
g) a genetic algorithm;

- 231 -

h) an annealing algorithm;
i) a weighted sum;
j) a nearest neighbor model; and,
k) a probabilistic model.
4. A method according to any one of the claims 1 to 3, wherein the method is
performed at
least in part using an electronic processing device.
5. A method according to any one of the claims 1 to 4, wherein the method
includes, in at
least one electronic processing device:
a) obtaining the pairs of biomarker values;
b) determining the first derived biomarker value;
c) determining the second derived biomarker value; and,
d) determining the indicator by adding the first and second derived biomarker
values.
6. A method according to any one of the claims 1 to 5, wherein the method
includes, in at
least one processing device, generating a representation of the indicator.
7. A method according to claim 6, wherein the representation includes:
a) an alphanumeric indication of the indicator;
b) a graphical indication of a comparison of the indicator to one or more
indicator
references;
c) an alphanumeric indication of a likelihood of the subject having at least
one medical
condition.
8. A method according to any one of the claims 1 to 7, wherein the method
includes:
a) comparing the indicator to an indicator reference; and,
b) determining a likelihood in accordance with results of the comparison.
9. A method according to claim 8, wherein the indicator reference is based on
at least one
of:
a) an indicator threshold range;
b) an indicator threshold; and,
c) an indicator distribution.
10. A method according to claim 8 or claim 9, wherein the indicator reference
is derived from
indicators determined for a number of individuals in a reference population.
11. A method according to claim 10, wherein the indicator reference is based
on a distribution
of indicators determined for a group of a reference population, the group
consisting of
individuals diagnosed as having the medical condition or lacking the medical
condition.

- 232 -

12. A method according to claim 10 or claim 11, wherein the reference
population includes:
a) a plurality of individuals of different sexes;
b) a plurality of individuals of different ethnicities;
c) a plurality of healthy individuals;
d) a plurality of individuals suffering from at least one diagnosed medical
condition;
e) a plurality of individuals lacking the at least one diagnosed medical
condition;
f) a plurality of individuals showing clinical signs of at least one
medical condition;
g) first and second groups of individuals, each group of individuals suffering
from a
respective diagnosed medical condition; and,
h) first and second groups of individuals, the first group of individuals
suffering from a
diagnosed medical condition, and the second group lacking the diagnosed
medical
condition.
13. A method according to any one of the claims 10 to 12, wherein the
indicator is for use in
determining the likelihood that a biological subject has at least one medical
condition, and
wherein the reference population includes:
a) individuals presenting with clinical signs of the at least one medical
condition;
b) individuals diagnosed as having the at least one medical condition;
c) individuals diagnosed as lacking the at least one medical condition; and,
d) healthy individuals.
14. A method according to any one of the claims 8 to 13, wherein the indicator
reference is
retrieved from a database.
15. A method according to any one of the claims 8 to 14, wherein the
likelihood is based on a
probability generated using the results of the comparison.
16. A method according any one of the claims 8 to 15, wherein the indicator is
for
determining a likelihood of the subject having a first or second condition,
and wherein the
method includes:
a) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of first and second conditions; and,
b) determining the likelihood in accordance with the results of the
comparison.
17. A method according to claim 16, wherein the method includes:
a) determining first and second indicator probabilities using the results of
the
comparisons; and,

- 233 -

b) combining the first and second indicator probabilities to determine a
condition
probability indicative of the likelihood.
18. A method according to claim 16 or claim 17, wherein the first and second
indicator
references are distributions of indicators determined for first and second
groups of a
reference population, the first and second group consisting of individuals
diagnosed with
the first or second condition respectively.
19. A method according to any one of the claims 1 to 18, wherein the method
includes:
a) obtaining a sample taken from the biological subject, the sample including
polynucleotide expression products; and,
b) quantifying at least some of the polynucleotide expression products within
the sample
to determine the pair of biomarker values.
20. A method according to claim 19, wherein the method includes, determining
the indicator
at least in part using a ratio of concentrations of the polynucleotide
expression products.
21. A method according to claim 19 or claim 20, wherein the method includes:
a) quantifying polynucleotide expression products by:
i) amplifying at least some polynucleotide expression products in the sample;
and,
ii) determining an amplification amount representing a degree of amplification

required to obtain a defined level of each of a pair of polynucleotide
expression
products; and,
b) determining the indicator by determining a difference between the
amplification
amounts.
22. A method according to claim 21, wherein the amplification amount is at
least one of:
a) a cycle time;
b) a number of cycles;
c) a cycle threshold;
d) an amplification time; and,
e) relative to an amplification amount of another amplified product.
23. A method according to claim 21 or claim 22, wherein the method includes
determining:
a) a first derived biomarker value by determining a difference between the
amplification
amounts of a first pair of polynucleotide expression products;
b) a second derived biomarker value by determining a difference between the
amplification amounts of a second pair of polynucleotide expression products;
c) determining the indicator by adding the first and second derived biomarker
values.

- 234 -

24. A method according to any one of the claims 1 to 23, wherein the immune
system
biomarker is a biomarker of an immune system of the biological subject that is
altered, or
whose level of expression is altered, as part of an inflammatory response to
damage or
pathogenic insult.
25. A method according to any one of the claims 1 to 24, wherein:
a) the at least two immune system biomarkers have a mutual correlation in
respect of the
at least one condition that lies within a mutual correlation range, the mutual

correlation range being between 0.9; and,
b) the indicator has a performance value greater than or equal to a
performance threshold
representing the ability of the indicator to diagnose the presence, absence,
degree or
prognosis of the at least one condition, the performance threshold being
indicative of
an explained variance of at least 0.3.
26. A method according to claim 25, wherein the mutual correlation range is at
least one of:
a) 0.8;
b) 0.7;
c) 0.6;
d) 0.5;
e) 0.4;
f) 0.3;
g) 0.2; and,
h) 0.1.
27. A method according to claim 25 or claim 26, wherein each immune system
biomarker has
a condition correlation with the presence, absence, degree or prognosis of the
at least one
condition that lies outside a condition correlation range, the condition
correlation range
being between 0.3 .
28. A method according to claim 27, wherein the condition correlation range is
at least one
of:
a) 0.9;
b) 0.8;
c) 0.7;
d) 0.6;
e) 0.5; and,
f) 0.4.

- 235 -

29. A method according to any one of the claims 25 to 28, wherein the
performance threshold
is indicative of an explained variance of at least one of:
a) 0.4;
b) 0.5;
c) 0.6;
d) 0.7;
e) 0.8; and,
f) 0.9.
30. A method according to any one of the claims 1 to 29, wherein the immune
system
biomarker value is indicative of a level or abundance of a molecule selected
from one or
more of a nucleic acid molecule and a proteinaceous molecule.
31. A method according to any one of the claims 1 to 30, wherein the indicator
is for
determining a likelihood of the subject having inSIRS or ipSIRS, and wherein
the method
includes:
a) determining a first pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the PLA2G7 gene and PLAC8 gene;
b) determining a second pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the CEACAM4 gene and LAMP 1 gene; and,
c) determining the indicator using the first and second pairs of biomarker
values.
32. A method according to any one of the claims 1 to 30, wherein the indicator
is for
determining a likelihood of the subject having inSIRS or a healthy condition,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group A IRS immune system biomarker genes

as herein defined; and
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group B IRS immune system biomarker genes

as herein defined.
33. A method according to any one of the claims 1 to 30, wherein the indicator
is for
determining a likelihood of the subject having ipSIRS or a healthy condition,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:

- 236 -

a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group C IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group D IRS immune system biomarker genes

as herein defined.
34. A method according to any one of the claims 1 to 30, wherein the indicator
is for
determining a likelihood of the subject having inSIRS or ipSIRS, and wherein
biomarker
values are determined from at least one immune system biomarker in each of
first and
second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group E IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group F IRS immune system biomarker genes

as herein defined.
35. A method according to any one of the claims 1 to 30, wherein the indicator
is for
determining a likelihood of the subject having inSIRS or ipSIRS, and wherein
biomarker
values are determined from at least one immune system biomarker in each of
first,
second, third and fourth IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group G IRS immune system biomarker genes

as herein defined;
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group H IRS immune system biomarker genes

as herein defined;
c) the third IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group I IRS immune system biomarker genes

as herein defined; and,
d) the fourth IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group J IRS immune system biomarker genes

as herein defined.

- 237 -

36. A method according to claim 35, wherein the first IRS immune system
biomarker is a
PLA2G7 expression product, wherein the second IRS immune system biomarker is a

PLAC8 expression product, wherein the third IRS immune system biomarker is a
CEACAM4 expression product and wherein the fourth IRS immune system biomarker
is a
LAMP1 expression product.
37. A method according to any one of the claims 1 to 30, wherein the indicator
is for
determining a likelihood of the subject having mild sepsis or severe sepsis,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group K IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group L IRS immune system biomarker genes

as herein defined.
38. A method according to any one of the claims 1 to 30, wherein the indicator
is for
determining a likelihood of the subject having mild sepsis or septic shock,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group M IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group N IRS immune system biomarker genes

as herein defined.
39. A method according to any one of the claims 1 to 30, wherein the indicator
is for
determining a likelihood of the subject having severe sepsis or septic shock,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group O IRS immune system biomarker genes

as herein defined; and,

- 238 -

b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group P IRS immune system biomarker genes

as herein defined.
40. Apparatus for determining an indicator used in assessing a likelihood of a
biological
subject having a presence, absence, degree or prognosis of at least one
medical condition,
the apparatus including at least one electronic processing device that:
a) determines a pair of biomarker values, each biomarker value being a value
measured
or derived for at least one corresponding immune system biomarker of the
biological
subject and being at least partially indicative of a concentration of the
immune system
biomarker in a sample taken from the subject;
b) determines a derived biomarker value using the pair of biomarker values,
the derived
biomarker value being indicative of a ratio of concentrations of the pair of
immune
system biomarkers; and,
c) determines the indicator using the derived biomarker value.
41. A composition comprising at least one pair of reverse transcribed mRNAs
and at least one
oligonucleotide primer or probe that hybridizes to an individual one of the
reverse
transcribed mRNAs, the at least one pair of reverse transcribed mRNAs
comprising a first
pair and a second pair of reverse transcribed mRNAs, wherein the first pair
comprises a
PLAC8 reverse transcribed mRNA and a PLA2G7 reverse transcribed mRNA and
wherein the second pair comprises a CEACAM4 reverse transcribed mRNA and a
LAMP 1
reverse transcribed mRNA.
42. A composition comprising at least one pair of reverse transcribed mRNAs
and at least one
oligonucleotide primer or probe that hybridizes to an individual one of the
reverse
transcribed mRNAs, the at least one pair of reverse transcribed mRNAs
comprising a
reverse transcribed mRNA from a first IRS immune system biomarker gene
selected from
group A IRS immune system biomarker genes as herein defined; and a reverse
transcribed
mRNA from a second IRS immune system biomarker gene selected from group B IRS
immune system biomarker genes as herein defined.
43. A composition comprising at least one pair of reverse transcribed mRNAs
and at least one
oligonucleotide primer or probe that hybridizes to an individual one of the
reverse
transcribed mRNAs, the at least one pair of reverse transcribed mRNAs
comprising a
reverse transcribed mRNA from a first IRS immune system biomarker gene
selected from
group C IRS immune system biomarker genes as herein defined; and a reverse
transcribed

- 239 -

mRNA from a second IRS immune system biomarker gene selected from group D IRS
immune system biomarker genes as herein defined.
44. A composition comprising at least one pair of reverse transcribed mRNAs
and at least one
oligonucleotide primer or probe that hybridizes to an individual one of the
reverse
transcribed mRNAs, the at least one pair of reverse transcribed mRNAs
comprising a
reverse transcribed mRNA from a first IRS immune system biomarker gene
selected from
group E IRS immune system biomarker genes as herein defined; and a reverse
transcribed
mRNA from a second IRS immune system biomarker gene selected from group F IRS
immune system biomarker genes as herein defined.
45. A composition comprising at least two pairs of reverse transcribed mRNAs
and at least
one oligonucleotide primer or probe that hybridizes to an individual one of
the reverse
transcribed mRNAs, the at least two pairs of reverse transcribed mRNAs
comprising a
first pair and a second pair of reverse transcribed mRNAs, wherein the first
pair
comprises a reverse transcribed mRNA from a first IRS immune system biomarker
gene
and a reverse transcribed mRNA from a second IRS immune system biomarker gene,
and
wherein the second pair comprises a reverse transcribed mRNA from a third IRS
immune
system biomarker gene and a reverse transcribed mRNA from a fourth IRS immune
system biomarker gene, wherein the first IRS immune system biomarker gene is
selected
from group G IRS immune system biomarker genes as herein defined, wherein the
second
IRS immune system biomarker gene is selected from group H IRS immune system
biomarker genes as herein defined, wherein the third IRS immune system
biomarker gene
is selected from group I IRS immune system biomarker genes as herein defined,
and
wherein the fourth IRS immune system biomarker gene is selected from group J
IRS
immune system biomarker genes as herein defined.
46. A composition comprising at least one pair of reverse transcribed mRNAs
and at least one
oligonucleotide primer or probe that hybridizes to an individual one of the
reverse
transcribed mRNAs, the at least one pair of reverse transcribed mRNAs
comprising a
reverse transcribed mRNA from a first IRS immune system biomarker gene
selected from
group K IRS immune system biomarker genes as herein defined; and a reverse
transcribed
mRNA from a second IRS immune system biomarker gene selected from group L IRS
immune system biomarker genes as herein defined.
47. A composition comprising at least one pair of reverse transcribed mRNAs
and at least one
oligonucleotide primer or probe that hybridizes to an individual one of the
reverse

- 240 -

transcribed mRNAs, the at least one pair of reverse transcribed mRNAs
comprising a
reverse transcribed mRNA from a first IRS immune system biomarker gene
selected from
group M IRS immune system biomarker genes as herein defined; and a reverse
transcribed mRNA from a second IRS immune system biomarker gene selected from
group N IRS immune system biomarker genes as herein defined.
48. A composition comprising at least one pair of reverse transcribed mRNAs
and at least one
oligonucleotide primer or probe that hybridizes to an individual one of the
reverse
transcribed mRNAs, the at least one pair of reverse transcribed mRNAs
comprising a
reverse transcribed mRNA from a first IRS immune system biomarker gene
selected from
group O IRS immune system biomarker genes as herein defined; and a reverse
transcribed
mRNA from a second IRS immune system biomarker gene selected from group P IRS
immune system biomarker genes as herein defined.
49. A composition according to any one of the claims 41 to 48, wherein the at
least one
oligonucleotide primer or probe is hybridized to an individual one of the
reverse
transcribed mRNAs.
50. A composition according to any one of the claims 41 to 48, wherein the
reverse
transcribed mRNAs are derived from components of the immune system.
51. A composition according to any one of the claims 41 to 48, wherein the
reverse
transcribed mRNAs are derived from leukocytes.
52. A composition according to any one of the claims 41 to 48, wherein the
reverse
transcribed mRNAs are derived from blood cells.
53. A composition according to any one of the claims 41 to 48, wherein the
reverse
transcribed mRNAs are derived from peripheral blood cells.
54. A composition according to any one of the claims 41 to 48, further
comprising a labeled
reagent for detecting the reverse transcribed mRNAs.
55. A composition according to claim 54, wherein the labeled reagent is a
labeled said at least
one oligonucleotide primer or probe.
56. A composition according to claim 54, wherein the labeled reagent is a
labeled said
reverse transcribed mRNA.
57. A kit for determining an indicator indicative of the likelihood of the
presence or absence
of at least one condition selected from the group consisting of inSIRS and
ipSIRS, the kit
comprising at least one pair of reagents comprising a first pair of reagents
and a second
pair of reagents, wherein the first pair of reagents comprises (i) a reagent
that allows

- 241 -

quantification of a polynucleotide expression product of the PLA2G7 gene; and
(ii) a
reagent that allows quantification of a polynucleotide expression product of
the PLAC8
gene, wherein the second pair of reagents comprises: (iii) a reagent that
allows
quantification of a polynucleotide expression product of the CEACAM4 gene; and
(iv) a
reagent that allows quantification of a polynucleotide expression product of
the LAMP1
gene.
58. A kit for determining an indicator indicative of the likelihood of the
presence or absence
of at least one condition selected from the group consisting of inSIRS and a
healthy
condition, the kit comprising at least one pair of reagents comprising (i) a
reagent that
allows quantification of a polynucleotide expression product of a first IRS
immune
system biomarker gene; and (ii) a reagent that allows quantification of a
polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS
immune system biomarker gene is selected from group A IRS immune system
biomarker
genes as herein defined, and wherein the second IRS immune system biomarker
gene is
selected from group B IRS immune system biomarker genes as herein defined.
59. A kit for determining an indicator indicative of the likelihood of the
presence or absence
of at least one condition selected from the group consisting of ipSIRS and a
healthy
condition, the kit comprising at least one pair of reagents comprising (i) a
reagent that
allows quantification of a polynucleotide expression product of a first IRS
immune
system biomarker gene; and (ii) a reagent that allows quantification of a
polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS
immune system biomarker gene is selected from group C IRS immune system
biomarker
genes as herein defined, and wherein the second IRS immune system biomarker
gene is
selected from group D IRS immune system biomarker genes as herein defined.
60. A kit for determining an indicator indicative of the likelihood of the
presence or absence
of at least one condition selected from the group consisting of inSIRS and
ipSIRS, the kit
comprising at least one pair of reagents comprising (i) a reagent that allows
quantification
of a polynucleotide expression product of a first IRS immune system biomarker
gene; and
(ii) a reagent that allows quantification of a polynucleotide expression
product of a second
IRS immune system biomarker gene, wherein the first IRS immune system
biomarker
gene is selected from group E IRS immune system biomarker genes as herein
defined,
and wherein the second IRS immune system biomarker gene is selected from group
F IRS
immune system biomarker genes as herein defined.

- 242 -

61. A kit for determining an indicator indicative of the likelihood of the
presence or absence
of at least one condition selected from the group consisting of inSIRS and
ipSIRS, the kit
comprising at least two pairs of reagents comprising a first pair of reagents
and a second
pair of reagents, wherein the first pair of reagents comprises (i) a reagent
that allows
quantification of a polynucleotide expression product of a first IRS immune
system
biomarker gene; and (ii) a reagent that allows quantification of a
polynucleotide
expression product of a second IRS immune system biomarker gene, and wherein
the
second pair of reagents comprises (i) a reagent that allows quantification of
a
polynucleotide expression product of a third IRS immune system biomarker gene;
and (ii)
a reagent that allows quantification of a polynucleotide expression product of
a fourth IRS
immune system biomarker gene, wherein the first IRS immune system biomarker
gene is
selected from group G IRS immune system biomarker genes as herein defined,
wherein
the second IRS immune system biomarker gene is selected from group H IRS
immune
system biomarker genes as herein defined, wherein the third IRS immune system
biomarker gene is selected from group I IRS immune system biomarker genes as
herein
defined, and wherein the fourth IRS immune system biomarker gene is selected
from
group J IRS immune system biomarker genes as herein defined.
62. A kit for determining an indicator indicative of the likelihood of the
presence or absence
of at least one condition selected from the group consisting of mild sepsis
and severe
sepsis, the kit comprising at least one pair of reagents comprising (i) a
reagent that allows
quantification of a polynucleotide expression product of a first IRS immune
system
biomarker gene; and (ii) a reagent that allows quantification of a
polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS
immune system biomarker gene is selected from group K IRS immune system
biomarker
genes as herein defined, and wherein the second IRS immune system biomarker
gene is
selected from group L IRS immune system biomarker genes as herein defined.
63. A kit for determining an indicator indicative of the likelihood of the
presence or absence
of at least one condition selected from the group consisting of mild sepsis
and septic
shock, the kit comprising at least one pair of reagents comprising (i) a
reagent that allows
quantification of a polynucleotide expression product of a first IRS immune
system
biomarker gene; and (ii) a reagent that allows quantification of a
polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS
immune system biomarker gene is selected from group M IRS immune system
biomarker

- 243 -

genes as herein defined, and wherein the second IRS immune system biomarker
gene is
selected from group N IRS immune system biomarker genes as herein defined.
64. A kit for determining an indicator indicative of the likelihood of the
presence or absence
of at least one condition selected from the group consisting of severe sepsis
and septic
shock, the kit comprising at least one pair of reagents comprising (i) a
reagent that allows
quantification of a polynucleotide expression product of a first IRS immune
system
biomarker gene; and (ii) a reagent that allows quantification of a
polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS
immune system biomarker gene is selected from group O IRS immune system
biomarker
genes as herein defined, and wherein the second IRS immune system biomarker
gene is
selected from group P IRS immune system biomarker genes as herein defined.
65. A method for inhibiting the development or progression in a subject of at
least one
condition selected from the group consisting of inSIRS and ipSIRS, the method
comprising: exposing the subject to a treatment regimen for treating the at
least one
condition based on an indicator obtained from an indicator-determining method,
wherein
the indicator is indicative of the presence of the at least one condition in
the subject, the
indicator-determining method comprising: (a) determining at least one pair of
biomarker
values, each biomarker value being a value measured or derived for at least
one
corresponding immune system biomarker of the biological subject and being at
least
partially indicative of a concentration of the immune system biomarker in a
sample taken
from the subject, (b) determining at least one derived biomarker value using
the at least
one pair of biomarker values, the derived biomarker value being indicative of
a ratio of
concentrations of the at least one pair of immune system biomarkers; and (c)
determining
the indicator based on the at least one derived biomarker value, wherein the
pair of
biomarker values comprises at least one of:
a) a first pair of biomarker values comprising first and second biomarker
values
corresponding to first and second biomarkers, wherein the first immune system
biomarker represents a polynucleotide expression product of the PLA2G7 gene
and
wherein the second immune system biomarker representing a polynucleotide
expression product of the PLAC8 gene, and
b) a second pair of biomarker values comprises third and fourth biomarker
values
corresponding to third and fourth immune system biomarkers, respectively,
wherein
the third immune system biomarker represents a polynucleotide expression
product of

- 244 -

the CEACAM4 gene and wherein the fourth immune system biomarker represents a
polynucleotide expression product of the LAMP1 gene.
66. A method according to claim 65, wherein the indicator-determining method
comprises:
determining the first pair and second pair of biomarker values and determining
a first
derived biomarker value calculated using the first pair of biomarker values
and a second
derived biomarker value calculated using the second pair of biomarker values;
and
determining the indicator based on a combination of the first and second
derived
biomarker values.
67. A method for inhibiting the development or progression of inSIRS in a
subject, the
method comprising: exposing the subject to a treatment regimen for treating
inSIRS based
on an indicator obtained from an indicator-determining method, wherein the
indicator is
indicative of the presence of inSIRS in the subject, the indicator-determining
method
comprising: (a) determining at least one pair of biomarker values, each
biomarker value
being a value measured or derived for at least one corresponding immune system

biomarker of the biological subject and being at least partially indicative of
a
concentration of the immune system biomarker in a sample taken from the
subject, (b)
determining at least one derived biomarker value using the at least one pair
of biomarker
values, the derived biomarker value being indicative of a ratio of
concentrations of the
pair of immune system biomarkers; and (c) determining the indicator based on
the at least
one derived biomarker value, wherein the at least one pair of biomarker values
comprises
first and second biomarker values corresponding to first and second immune
system
biomarkers, respectively, wherein the first immune system biomarker represents
a
polynucleotide expression product of a first IRS immune system biomarker gene,
and
wherein the second immune system biomarker represents a polynucleotide
expression
product of a second IRS immune system biomarker gene, wherein the first IRS
immune
system biomarker gene is selected from group A IRS immune system biomarker
genes as
herein defined; and wherein the second IRS immune system biomarker is selected
from
group B IRS immune system biomarker genes as herein defined.
68. A method for inhibiting the development or progression of ipSIRS in a
subject, the
method comprising: exposing the subject to a treatment regimen for treating
ipSIRS based
on an indicator obtained from an indicator-determining method, wherein the
indicator is
indicative of the presence of ipSIRS in the subject, the indicator-determining
method
comprising: (a) determining at least one pair of biomarker values, each
biomarker value

- 245 -

being a value measured or derived for at least one corresponding immune system

biomarker of the biological subject and being at least partially indicative of
a
concentration of the immune system biomarker in a sample taken from the
subject, (b)
determining at least one derived biomarker value using the at least one pair
of biomarker
values, the derived biomarker value being indicative of a ratio of
concentrations of the at
least one pair of immune system biomarkers; and (c) determining the indicator
based on
the at least one derived biomarker value, wherein the at least one pair of
biomarker values
comprises first and second biomarker values corresponding to first and second
immune
system biomarkers, respectively, wherein the first immune system biomarker
represents a
polynucleotide expression product of a first IRS immune system biomarker gene,
and
wherein the second immune system biomarker represents a polynucleotide
expression
product of a second IRS immune system biomarker gene, wherein the first IRS
immune
system biomarker gene is selected from group C IRS immune system biomarker
genes as
herein defined; and wherein the second IRS immune system biomarker is selected
from
group D IRS immune system biomarker genes as herein defined.
69. A method for inhibiting the development or progression in a subject of at
least one
condition selected from the group consisting of inSIRS and ipSIRS, the method
comprising: exposing the subject to a treatment regimen for treating the at
least one
condition based on an indicator obtained from an indicator-determining method,
wherein
the indicator is indicative of the presence of the at least one condition in
the subject, the
indicator-determining method comprising: (a) determining at least one pair of
biomarker
values, each biomarker value being a value measured or derived for at least
one
corresponding immune system biomarker of the biological subject and being at
least
partially indicative of a concentration of the immune system biomarker in a
sample taken
from the subject, (b) determining at least one derived biomarker value using
the at least
one pair of biomarker values, the derived biomarker value being indicative of
a ratio of
concentrations of the at least one pair of immune system biomarkers; and (c)
determining
the indicator based on the at least one derived biomarker value, wherein the
at least one
pair of biomarker values comprises first and second biomarker values
corresponding to of
first and second immune system biomarkers, respectively, wherein the first
immune
system biomarker represents a polynucleotide expression product of a first IRS
immune
system biomarker gene, and wherein the second immune system biomarker
represents a
polynucleotide expression product of a second IRS immune system biomarker
gene,

- 246 -
wherein the first IRS immune system biomarker gene is selected from group E
IRS
immune system biomarker genes as herein defined; and wherein the second IRS
immune
system biomarker is selected from group F IRS immune system biomarker genes as

herein defined.
70. A method for inhibiting the development or progression in a subject of at
least one
condition selected from the group consisting of inSIRS and ipSIRS, the method
comprising: exposing the subject to a treatment regimen for treating the at
least one
condition based on an indicator obtained from an indicator-determining method,
wherein
the indicator is indicative of the presence of the at least one condition in
the subject, the
indicator-determining method comprising: (a) determining at least two pairs of
biomarker
values, each biomarker value being a value measured or derived for at least
one
corresponding immune system biomarker of the biological subject and being at
least
partially indicative of a concentration of the immune system biomarker in a
sample taken
from the subject, (b) determining at least two derived biomarker values using
the at least
two pairs of biomarker values, the derived biomarker value being indicative of
a ratio of
concentrations of each pair of immune system biomarkers; and (c) determining
the
indicator based on the at least two derived biomarker values, wherein the at
least one pair
of biomarker values comprises a first pair of biomarker values comprising
first and
second biomarker values corresponding to first and second immune system
biomarkers,
respectively, wherein the first immune system biomarker represents a
polynucleotide
expression product of a first IRS immune system biomarker gene and wherein the
second
immune system biomarker represents a polynucleotide expression product of a
second
IRS immune system biomarker gene, and a second pair of biomarker values
comprising
third and fourth biomarker values corresponding to third and fourth immune
system
biomarkers, respectively, wherein the third immune system biomarker represents
a
polynucleotide expression product of a third IRS immune system biomarker gene
and
wherein the fourth immune system biomarker represents a polynucleotide
expression
product of a fourth IRS immune system biomarker gene, wherein the first IRS
immune
system biomarker gene is selected from group G IRS immune system biomarker
genes as
herein defined, wherein the second IRS immune system biomarker gene is
selected from
group H IRS immune system biomarker genes as herein defined, wherein the third
IRS
immune system biomarker gene is selected from group I IRS immune system
biomarker

- 247 -
genes as herein defined, and wherein the fourth IRS immune system biomarker
gene is
selected from group J IRS immune system biomarker genes as herein defined.
71. A method for inhibiting the development or progression in a subject of at
least one
condition selected from the group consisting of mild sepsis and severe sepsis,
the method
comprising: exposing the subject to a treatment regimen for treating the at
least one
condition based on an indicator obtained from an indicator-determining method,
wherein
the indicator is indicative of the presence of the at least one condition in
the subject, the
indicator-determining method comprising: (a) determining at least one pair of
biomarker
values, each biomarker value being a value measured or derived for at least
one
corresponding immune system biomarker of the biological subject and being at
least
partially indicative of a concentration of the immune system biomarker in a
sample taken
from the subject, (b) determining at least one derived biomarker value using
the at least
one pair of biomarker values, the derived biomarker value being indicative of
a ratio of
concentrations of the at least one pair of immune system biomarkers; and (c)
determining
the indicator based on the at least one derived biomarker value, wherein the
at least one
pair of biomarker values comprises first and second biomarker values
corresponding to
first and second immune system biomarkers, respectively, wherein the first
immune
system biomarker represents a polynucleotide expression product of a first IRS
immune
system biomarker gene, and wherein the second immune system biomarker
represents a
polynucleotide expression product of a second IRS immune system biomarker
gene,
wherein the first IRS immune system biomarker gene is selected from group K
IRS
immune system biomarker genes as herein defined; and wherein the second IRS
immune
system biomarker is selected from group L IRS immune system biomarker genes as

herein defined.
72. A method for inhibiting the development or progression in a subject of at
least one
condition selected from the group consisting of mild sepsis and septic shock,
the method
comprising: exposing the subject to a treatment regimen for treating the at
least one
condition based on an indicator obtained from an indicator-determining method,
wherein
the indicator is indicative of the presence of the at least one condition in
the subject, the
indicator-determining method comprising: (a) determining at least one pair of
biomarker
values, each biomarker value being a value measured or derived for at least
one
corresponding immune system biomarker of the biological subject and being at
least
partially indicative of a concentration of the immune system biomarker in a
sample taken

- 248 -
from the subject, (b) determining at least one derived biomarker value using
the at least
one pair of biomarker values, the derived biomarker value being indicative of
a ratio of
concentrations of the at least one pair of immune system biomarkers; and (c)
determining
the indicator based on the at least one derived biomarker value, wherein the
at least one
pair of biomarker values comprises first and second biomarker values
corresponding to
first and second immune system biomarkers, respectively, wherein the first
immune
system biomarker represents a polynucleotide expression product of a first IRS
immune
system biomarker gene, and wherein the second immune system biomarker
represents a
polynucleotide expression product of a second IRS immune system biomarker
gene,
wherein the first IRS immune system biomarker gene is selected from group M
IRS
immune system biomarker genes as herein defined; and wherein the second IRS
immune
system biomarker is selected from group N IRS immune system biomarker genes as

herein defined.
73. A method for inhibiting the development or progression in a subject of at
least one
condition selected from the group consisting of severe sepsis and septic
shock, the
method comprising: exposing the subject to a treatment regimen for treating
the at least
one condition based on an indicator obtained from an indicator-determining
method,
wherein the indicator is indicative of the presence of the at least one
condition in the
subject, the indicator-determining method comprising: (a) determining at least
one pair of
biomarker values, each biomarker value being a value measured or derived for
at least
one corresponding immune system biomarker of the biological subject and being
at least
partially indicative of a concentration of the immune system biomarker in a
sample taken
from the subject, (b) determining at least one derived biomarker value using
the at least
one pair of biomarker values, the derived biomarker value being indicative of
a ratio of
concentrations of the at least one pair of immune system biomarkers; and (c)
determining
the indicator based on the at least one derived biomarker value, wherein the
at least one
pair of biomarker values comprises first and second biomarker values
corresponding to
first and second immune system biomarkers, respectively, wherein the first
immune
system biomarker represents a polynucleotide expression product of a first IRS
immune
system biomarker gene, and wherein the second immune system biomarker
represents a
polynucleotide expression product of a second IRS immune system biomarker
gene,
wherein the first IRS immune system biomarker gene is selected from group O
IRS
immune system biomarker genes as herein defined; and wherein the second IRS
immune

- 249 -
system biomarker is selected from group P IRS immune system biomarker genes as

herein defined.
74. A method according to any one of claims 65 to 73, comprising: taking the
sample from
the subject and obtaining the indicator according to the indicator-determining
method.
75. A method according to any one of claims 65 to 73, comprising: sending the
sample taken
from the subject to a laboratory at which the indicator is determined.
76. A method for differentiating between inSIRS and ipSIRS in a biological
subject, the
method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a pair
of biomarker values, the pair of biomarker values being selected from the
group
consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the PLA2G7 gene and PLAC8 gene;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the CEACAM4 gene and LAMP 1 gene;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide
expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of inSIRS and ipSIRS, respectively; and,
e) determining a likelihood of the subject having inSIRS or ipSIRS in
accordance with
the results of the comparison.
77. A method for differentiating between inSIRS and a healthy condition in a
biological
subject, the method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a pair
of biomarker values, the pair of biomarker values being indicative of a
concentration
of polynucleotide expression products of a first IRS immune system biomarker
gene
and a second IRS immune system biomarker gene, wherein the first IRS immune
system biomarker gene is selected from group A IRS immune system biomarker

- 250 -
genes as herein defined, and wherein the second IRS immune system biomarker
gene
is selected from group B IRS immune system biomarker genes as herein defined;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide
expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of inSIRS and healthy condition,
respectively;
and,
e) determining a likelihood of the subject having inSIRS or the healthy
condition in
accordance with the results of the comparison.
78. A method for differentiating between ipSIRS and a healthy condition in a
biological
subject, the method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a pair
of biomarker values, the pair of biomarker values being indicative of a
concentration
of polynucleotide expression products of a first IRS immune system biomarker
gene
and a second IRS immune system biomarker gene, wherein the first IRS immune
system biomarker gene is selected from group C IRS immune system biomarker
genes as herein defined and wherein the second IRS immune system biomarker
gene
is selected from group D IRS immune system biomarker genes as herein defined;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide
expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of ipSIRS and healthy condition,
respectively;
and,
e) determining a likelihood of the subject having ipSIRS or the healthy
condition in
accordance with the results of the comparison.
79. A method for differentiating between inSIRS and ipSIRS in a biological
subject, the
method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a pair
of biomarker values, the pair of biomarker values being indicative of a
concentration

- 251 -
of polynucleotide expression products of a first IRS immune system biomarker
gene
and a second IRS immune system biomarker gene, wherein the first IRS immune
system biomarker gene is selected from group E IRS immune system biomarker
genes
as herein defined, and wherein the second IRS immune system biomarker gene is
selected from group F IRS immune system biomarker genes as herein defined;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide
expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of inSIRS and ipSIRS, respectively; and,
e) determining a likelihood of the subject having inSIRS or ipSIRS in
accordance with
the results of the comparison.
80. A method for differentiating between inSIRS and ipSIRS in a biological
subject, the
method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a pair
of biomarker values, the pair of biomarker values being selected from the
group
consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of a first IRS immune system biomarker gene and a second
IRS immune system biomarker gene, wherein the first IRS immune system
biomarker gene is selected from group G IRS immune system biomarker genes as
herein defined, and wherein the second IRS immune system biomarker gene is
selected from group H IRS immune system biomarker genes as herein defined;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide
expression products of a third IRS immune system biomarker gene and a fourth
IRS immune system biomarker gene, wherein the third IRS immune system
biomarker gene is selected from group I IRS immune system biomarker genes as
herein defined, and wherein the fourth IRS immune system biomarker gene is
selected from group J IRS immune system biomarker genes as herein defined;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide
expression products using the pair of biomarker values; and,

- 252 -
d) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of inSIRS and ipSIRS, respectively; and,
e) determining a likelihood of the subject having inSIRS or ipSIRS in
accordance with
the results of the comparison.
81. A method for differentiating between mild sepsis and severe sepsis in a
biological subject,
the method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a pair
of biomarker values, the pair of biomarker values being indicative of a
concentration
of polynucleotide expression products of a first IRS immune system biomarker
gene
and a second IRS immune system biomarker gene, wherein the first IRS immune
system biomarker gene is selected from group K IRS immune system biomarker
genes as herein defined, and wherein the second IRS immune system biomarker
gene
is selected from group L IRS immune system biomarker genes as herein defined;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide
expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of mild sepsis and severe sepsis,
respectively;
and,
e) determining a likelihood of the subject having mild sepsis or severe sepsis
in
accordance with the results of the comparison.
82. A method for differentiating between mild sepsis and septic shock in a
biological subject,
the method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a pair
of biomarker values, the pair of biomarker values being indicative of a
concentration
of polynucleotide expression products of a first IRS immune system biomarker
gene
and a second IRS immune system biomarker gene, wherein the first IRS immune
system biomarker gene is selected from group M IRS immune system biomarker
genes as herein defined, and wherein the second IRS immune system biomarker
gene
is selected from group N IRS immune system biomarker genes as herein defined;

- 253 -
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide
expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of mild sepsis and septic shock,
respectively;
and,
e) determining a likelihood of the subject having mild sepsis or septic shock
in
accordance with the results of the comparison.
83. A method for differentiating between severe sepsis and septic shock in a
biological
subject, the method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a pair
of biomarker values, the pair of biomarker values being indicative of a
concentration
of polynucleotide expression products of a first IRS immune system biomarker
gene
and a second IRS immune system biomarker gene, wherein the first IRS immune
system biomarker gene is selected from group O IRS immune system biomarker
genes as herein defined, and wherein the second IRS immune system biomarker
gene
is selected from group P IRS immune system biomarker genes as herein defined;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide
expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of severe sepsis and septic shock,
respectively;
and,
e) determining a likelihood of the subject having severe sepsis or septic
shock in
accordance with the results of the comparison.
84. A method according to claim 76 or claim 80, wherein the method includes:
a) determining a first derived biomarker using the first pair of biomarker
values;
b) determining a second derived biomarker value using the second pair of
biomarker
values; and,
c) determining the indicator by combining the first and second derived
biomarker
values.
85. A method according to any one of claims 76 to 84, wherein the first and
second indicator
references are distributions of indicators determined for first and second
groups of a

- 254 -
reference population, the first and second group consisting of individuals
diagnosed with
inSIRS and ipSIRS respectively.
86. A method for determining an indicator used in assessing the likelihood of
a biological
subject having at least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required
to obtain a defined level of each of a pair of polynucleotide expression
products
selected from the group consisting of:
i) a first pair of polynucleotide expression products of the PLA2G7
gene and PLAC8
gene;
ii) a second pair of polynucleotide expression products of the CEACAM4 gene
and
LAMP 1 gene;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a medical
condition.
87. A method for determining an indicator used in assessing the likelihood of
a biological
subject having at least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required
to obtain a defined level of each of a pair of polynucleotide expression
products
selected from the group consisting of: a polynucleotide expression product of
a first
IRS immune system biomarker gene and a polynucleotide expression product of a
second IRS immune system biomarker gene, wherein the first IRS immune system
biomarker gene is selected from group A IRS immune system biomarker genes as
herein defined, and wherein the second IRS immune system biomarker gene is
selected from group B IRS immune system biomarker genes as herein defined;
d) determining the indicator by determining a difference between the
amplification
amounts; and,

- 255 -
e) using the indicator to assess the likelihood of a biological subject having
a medical
condition.
88. A method for determining an indicator used in assessing the likelihood of
a biological
subject having at least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required
to obtain a defined level of each of a pair of polynucleotide expression
products
selected from the group consisting of: a polynucleotide expression product of
a first
IRS immune system biomarker gene and a polynucleotide expression product of a
second IRS immune system biomarker gene, wherein the first IRS immune system
biomarker gene is selected from group C IRS immune system biomarker genes as
herein defined, and wherein the second IRS immune system biomarker gene is
selected from group D IRS immune system biomarker genes as herein defined;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a medical
condition.
89. A method for determining an indicator used in assessing the likelihood of
a biological
subject having at least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required
to obtain a defined level of each of a pair of polynucleotide expression
products
selected from the group consisting of: a polynucleotide expression product of
a first
IRS immune system biomarker gene and a polynucleotide expression product of a
second IRS immune system biomarker gene, wherein the first IRS immune system
biomarker gene is selected from group E IRS immune system biomarker genes as
herein defined, and wherein the second IRS immune system biomarker gene is
selected from group F IRS immune system biomarker genes as herein defined;

- 256 -
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a medical
condition.
90. A method for determining an indicator used in assessing the likelihood of
a biological
subject having at least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required
to obtain a defined level of each of a pair of polynucleotide expression
products
selected from the group consisting of:
i) a first pair of polynucleotide expression products of a first IRS immune
system
biomarker gene and a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group G IRS immune
system biomarker genes as herein defined, and wherein the second IRS immune
system biomarker gene is selected from group H IRS immune system biomarker
genes as herein defined;
ii) a second pair of polynucleotide expression products of a third IRS immune
system
biomarker gene and a fourth IRS immune system biomarker gene, wherein the
third IRS immune system biomarker gene is selected from group I IRS immune
system biomarker genes as herein defined, and wherein the fourth IRS immune
system biomarker gene is selected from group J IRS immune system biomarker
genes as herein defined;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a medical
condition.
91. A method for determining an indicator used in assessing the likelihood of
a biological
subject having at least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;

- 257 -
c) determining an amplification amount representing a degree of amplification
required
to obtain a defined level of each of a pair of polynucleotide expression
products
selected from the group consisting of: a polynucleotide expression product of
a first
IRS immune system biomarker gene and a polynucleotide expression product of a
second IRS immune system biomarker gene, wherein the first IRS immune system
biomarker gene is selected from group K IRS immune system biomarker genes as
herein defined, and wherein the second IRS immune system biomarker gene is
selected from group L IRS immune system biomarker genes as herein defined;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a medical
condition.
92. A method for determining an indicator used in assessing the likelihood of
a biological
subject having at least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required
to obtain a defined level of each of a pair of polynucleotide expression
products
selected from the group consisting of: a polynucleotide expression product of
a first
IRS immune system biomarker gene and a polynucleotide expression product of a
second IRS immune system biomarker gene,, wherein the first IRS immune system
biomarker gene is selected from group M IRS immune system biomarker genes as
herein defined, and wherein the second IRS immune system biomarker gene is
selected from group N IRS immune system biomarker genes as herein defined;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a medical
condition.
93. A method for determining an indicator used in assessing the likelihood of
a biological
subject having at least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;

- 258 -
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required
to obtain a defined level of each of a pair of polynucleotide expression
products
selected from the group consisting of: a polynucleotide expression product of
a first
IRS immune system biomarker gene and a polynucleotide expression product of a
second IRS immune system biomarker gene, wherein the first IRS immune system
biomarker gene is selected from group O IRS immune system biomarker genes as
herein defined, and wherein the second IRS immune system biomarker gene is
selected from group P IRS immune system biomarker genes as herein defined;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a medical
condition.
94. A method according to any one of the claims 86 to 93, wherein the method
includes
determining:
a) a first derived biomarker value by determining a difference between the
amplification
amounts of the first pair of polynucleotide expression products;
b) a second derived biomarker value by determining a difference between the
amplification amounts of the second pair of polynucleotide expression
products;
c) determining the indicator by adding the first and second derived biomarker
values.
95. A method according to any one of the claims 86 to 94, wherein the method
includes
determining:
a) comparing the indicator to first and second indicator references, wherein
the first and
second indicator references are distributions of indicators determined for
first and
second groups of a reference population, one of the first and second groups
consisting
of individuals diagnosed with the medical condition; and,
b) determining a likelihood of the subject having the medical condition in
accordance
with the results of the comparison.
96. A method according to any one of claims 76 to 95, wherein the
amplification amount is at
least one of:
a) a cycle time;
b) a number of cycles;
c) a cycle threshold;

- 259 -
d) an amplification time; and,
e) relative to an amplification amount of another amplified product.
97. A method for use in assessing the likelihood of a biological subject
having a medical
condition, the method including, in one or more processing devices:
a) determining a pair of biomarker values, the pair of biomarker values being
selected
from the group consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the PLA2G7 gene and PLAC8 gene;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the CEACAM4 gene and LAMP1 gene;
b) determining an indicator indicative of a ratio of the concentrations of the

polynucleotide expression products using the pair of biomarker values;
c) retrieving previously determined first and second indicator references from
a
database, the first and second indicator references being determined based on
indicators determined from first and second groups of a reference population,
one of
the groups consisting of individuals diagnosed with the medical condition;
d) comparing the indicator to the first and second indicator references;
e) using the results of the comparison to determine a probability indicative
of the subject
having the medical condition; and,
f) generating a representation of the probability, the representation being
displayed to a
user to allow the user to assess the likelihood of a biological subject having
at least
one medical condition.
98. A method according to claim 97, wherein the method includes:
a) determining a first derived biomarker value using the first pair of
biomarker values;
b) determining a second derived biomarker value using the second pair of
biomarker
values; and,
c) determining the indicator by combining the first and second derived
biomarker
values.
99. Apparatus for determining an indicator used in determining the likelihood
of a biological
subject having at least one medical condition, the apparatus including:
a) a sampling device that obtains a sample taken from a biological subject,
the sample
including polynucleotide expression products;

- 260 -
b) a measuring device that quantifies polynucleotide expression products
within the
sample to determine a pair of biomarker values, the pair of biomarker values
being
selected from the group consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the PLA2G7 gene and PLAC8 gene;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the CEACAM4 gene and LAMP1 gene;
c) at least one processing device that:
i) receives an indication of the pair of biomarker values from the
measuring device;
ii) determines an indicator using a ratio of the concentration of the first
and second
polynucleotide expression products using the biomarker values; and,
iii) compares the indicator to at least one indicator reference; and,
iv) determines a likelihood of the subject having the at least one medical
condition
using the results of the comparison; and,
v) generates a representation of the indicator and the likelihood for display
to a user.
100. A method for differentiating between inSIRS and ipSIRS in a biological
subject, the
method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of SIRS,
the sample including polynucleotide expression products;
b) in a measuring device:
i) amplifying at least some polynucleotide expression products in the
sample;
ii) determining an amplification amount representing a degree of amplification

required to obtain a defined level of polynucleotide expression products
including:
(1) amplification amounts for a first pair of polynucleotide expression
products of
the PLA2G7 gene and PLAC8 gene;
(2) amplification amounts for a second pair of polynucleotide expression
products
of the CEACAM4 gene and LAMP1 gene;
c) in a processing system:
i) retrieving the amplification amounts;
ii) determining an indicator by:
(1) determining a first derived biomarker value indicative of a ratio of
concentrations of the first pair of polynucleotide expression products by
determining a difference between the amplification amounts for the first pair;

- 261 -
(2) determining a second derived biomarker value indicative of a ratio of
concentrations of the second pair of polynucleotide expression products by
determining a difference between the amplification amounts for the second
pair;
(3) determining the indicator by adding the first and second derived biomarker

values;
iii) retrieving previously determined first and second indicator references
from a
database, wherein the first and second indicator references are distributions
of
indicators determined for first and second groups of a reference population,
the
first and second group consisting of individuals diagnosed with inSIRS and

respectively;
iv) comparing the indicator to the first and second indicator references;
v) using the results of the comparison to determine a probability of the
subject being
classified within the first or second group;
vi) generating a representation at least partially indicative of the indicator
and the
probability; and,
vii)providing the representation to a user to allow the user to assess the
likelihood of
a biological subject having at least one medical condition.
101. A method for determining an indicator used in assessing a likelihood of a
biological
subject having a presence, absence, degree or prognosis of at least one
medical condition,
the method including:
a) determining a plurality of biomarker values, each biomarker value being
indicative of
a value measured or derived for at least one corresponding immune system
biomarker
of the biological subject and being at least partially indicative of a
concentration of
the immune system biomarker in a sample taken from the subject;
b) determining the indicator using a combination of the plurality of biomarker
values,
wherein:
i) at least two biomarkers have a mutual correlation in respect of the at
least one
condition that lies within a mutual correlation range, the mutual correlation
range
being between ~0.9; and,
ii) the indicator has a performance value greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,

- 262 -
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.
102. A method according to claim 101, wherein the method includes:
a) determining a plurality of measured biomarker values, each measured
biomarker
value being a measured value of a corresponding biomarker of the biological
subject;
and,
b) determining the indicator by applying a function to at least one of the
measured
biomarker values to determine at least one derived biomarker value, the at
least one
derived biomarker value being indicative of a value of a corresponding derived

biomarker.
103. A method according to claim 102, wherein the function includes at least
one of:
a) multiplying two biomarker values;
b) dividing two biomarker values;
c) adding two biomarker values;
d) subtracting two biomarker values;
e) a ratio of two biomarker values;
f) a weighted sum of at least two biomarker values;
g) a log sum of at least two biomarker values; and,
h) a sigmoidal function of at least two biomarker values.
104. A method according to claim 102, wherein the method includes determining
at least
one derived biomarker value corresponding to a ratio of two measured biomarker
values.
105. A method according to any one of the claims 101to 104, wherein the method
includes
combining at least two biomarker values to determine an indicator value
representing the
indicator.
106. A method according to claim 105, wherein the method includes combining at
least
two biomarker values using a combining function, the combining function being
at least
one of:
a) an additive model;
b) a linear model;
c) a support vector machine;
d) a neural network model;
e) a random forest model;
f) a regression model;

- 263 -
g) a genetic algorithm;
h) an annealing algorithm;
i) a weighted sum;
j) a nearest neighbor model; and,
k) a probabilistic model.
107. A method according to claim 105 or claim 106, wherein at least one of the
at least two
biomarkers is a derived biomarker.
108. A method according to any one of the claims 101 to 107, wherein the
method
includes:
a) determining a first derived biomarker value, the first derived biomarker
value being
indicative of a ratio of concentrations of the first and second immune system
biomarkers;
b) determining a second derived biomarker value, the second derived biomarker
value
being indicative of a ratio of concentrations of the third and fourth measured
immune
system biomarkers; and,
c) adding the first and second derived biomarker values to generate an
indicator value.
109. A method according to any one of the claims 101 to 108, wherein the
method is
performed at least in part using an electronic processing device.
110. A method according to any one of the claims 101 to 109, wherein the
method
includes, in the electronic processing device:
a) receiving a plurality of measured biomarker values, each measured biomarker
value
being a measured value of a corresponding immune system biomarker;
b) applying a function to at least one of the measured biomarker values to
determine at
least one derived biomarker value, the at least one derived biomarker value
being
indicative of a value of a corresponding derived biomarker; and,
c) combining at least one derived biomarker value and at least one other
biomarker
value to determine the indicator.
111. A method according to any one of the claims 101 to 110, wherein the
mutual
correlation range is at least one of:
a) ~0.8;
b) ~0.7;
c) ~0.6;
d) ~0.5;

- 264 -
e) ~0.4;
f) ~0.3;
g) ~0.2; and,
h) ~0.1.
112. A method according to any one of the claims 101 to 111, wherein each
biomarker has
a condition correlation with the presence, absence, degree or prognosis of the
at least one
condition that lies outside a condition correlation range, the condition
correlation range
being between ~0.3 .
113. A method according to claim 112, wherein the condition correlation range
is at least
one of:
a) ~0.9;
b) ~0.8;
c) ~0.7;
d) ~0.6;
e) ~0.5; and,
f) ~0.4.
114. A method according to any one of the claims 101 to 113, wherein the
performance
threshold is indicative of an explained variance of at least one of:
a) 0.4;
b) 0.5;
c) 0.6;
d) 0.7;
e) 0.8; and,
f) 0.9.
115. A method according to any one of the claims 101 to 114, wherein the
biomarker value
is indicative of a level or abundance of a molecule selected from one or more
of a nucleic
acid molecule and a proteinaceous molecule.
116. A method according to any one of the claims 101 to 115, wherein the
method includes
generating a representation of the indicator.
117. A method according to claim 116, wherein the representation includes:
a) an alphanumeric indication of the indicator;
b) a graphical indication of a comparison of the indicator to one or more
indicator
references;

- 265 -
c) an alphanumeric indication of a likelihood of the subject having at least
one medical
condition.
118. A method according to any one of the claims 101 to 117, wherein the
method
includes:
a) comparing the indicator to an indicator reference; and,
b) determining a likelihood in accordance with results of the comparison.
119. A method according to claim 118, wherein the indicator reference is based
on at least
one of:
a) an indicator threshold range;
b) an indicator threshold; and,
c) an indicator distribution.
120. A method according to claim 118 or claim 119, wherein the indicator
reference is
derived from indicators determined for a number of individuals in a reference
population.
121. A method according to claim 120, wherein the indicator reference is based
on a
distribution of indicators determined for a group of a reference population,
the group
consisting of individuals diagnosed as having the medical condition or lacking
the
medical condition.
122. A method according to claim 120 or claim 121, wherein the reference
population
includes:
a) a plurality of individuals of different sexes;
b) a plurality of individuals of different ethnicities;
c) a plurality of healthy individuals;
d) a plurality of individuals suffering from at least one diagnosed medical
condition;
e) a plurality of individuals without the at least one diagnosed medical
condition;
f) a plurality of individuals showing clinical signs of at least one
medical condition;
g) first and second groups of individuals, each group of individuals suffering
from a
respective diagnosed medical condition; and,
h) first and second groups of individuals, the first group of individuals
suffering from a
diagnosed medical condition, and the second group not suffering the diagnosed
medical condition.
123. A method according to any one of the claims 120 to 122, wherein the
indicator is for
use in determining the likelihood that a biological subject has at least one
medical
condition, and wherein the reference population includes:

- 266 -
a) individuals presenting with clinical signs of the at least one medical
condition;
b) individuals diagnosed with the at least one medical condition;
c) individuals diagnosed without the at least one medical condition; and,
d) healthy individuals.
124. A method according to any one of the claims 118 to 123, wherein the
indicator
reference is retrieved from a database.
125. A method according to any one of the claims 118 to 124, wherein the
likelihood is
based on a probability generated using the results of the comparison.
126. A method according any one of the claims 118 to 125, wherein the
indicator is for
determining a likelihood of the subject having a first or second condition,
and wherein the
method includes:
a) comparing the indicator to first and second indicator references, the first
and second
indicator references being indicative of first and second conditions; and,
b) determining the likelihood in accordance with the results of the
comparison.
127. A method according to claim 126, wherein the method includes:
a) determining first and second indicator probabilities using the results of
the
comparisons; and,
b) combining the first and second indicator probabilities to determine a
condition
probability indicative of the likelihood.
128. A method according to claim 126 or claim 127, wherein the first and
second indicator
references are distributions of indicators determined for first and second
groups of a
reference population, the first and second group consisting of individuals
diagnosed with
the first or second condition respectively.
129. A method according to any one of the claims 101 to 128, wherein the
method
includes:
a) obtaining a sample taken from the biological subject, the sample including
polynucleotide expression products;
b) quantifying at least some of the polynucleotide expression products within
the sample
to determine at least a pair of biomarker values;
c) determining the indicator at least in part using the pair of biomarker
values;
130. A method according to claim 129, wherein the method includes,
determining the
indicator at least in part using a ratio of concentrations of the
polynucleotide expression
products.

- 267 -
131. A method according to claim 129 or claim 130, wherein the method
includes:
a) quantifying polynucleotide expression products by:
i) amplifying at least some polynucleotide expression products in the
sample; and,
ii) determining an amplification amount representing a degree of amplification

required to obtain a defined level of each of a pair of polynucleotide
expression
products; and,
b) determining the indicator by determining a difference between the
amplification
amounts.
132. A method according to claim 131, wherein the amplification amount is at
least one of:
a) a cycle time;
b) a number of cycles;
c) a cycle threshold;
d) an amplification time; and,
e) relative to an amplification amount of another amplified product.
133. A method according to claim 131 or claim 132, wherein the method includes

determining:
a) a first derived biomarker value by determining a difference between the
amplification
amounts of a first pair of polynucleotide expression products;
b) a second derived biomarker value by determining a difference between the
amplification amounts of a second pair of polynucleotide expression products;
c) determining the indicator by adding the first and second derived biomarker
values.
134. A method according to any one of the claims 101 to 133, wherein the
immune system
biomarker is a biomarker of an immune system of the biological subject that is
altered, or
whose level of expression is altered, as part of an inflammatory response to
damage or
pathogenic insult.
135. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having at least one of inSIRS and
ipSIRS, and
wherein the method includes:
a) determining a first pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the PLA2G7 gene and PLAC8 gene;
b) determining a second pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the CEACAM4 gene and LAMP1 gene; and,
c) determining the indicator using the first and second pairs of biomarker
values.

- 268 -
136. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having inSIRS or ipSIRS, and wherein
the method
includes:
a) determining a first pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the PLA2G7 gene and PLAC8 gene;
b) determining a second pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the CEACAM4 gene and LAMP1 gene; and,
c) determining the indicator using the first and second pairs of biomarker
values.
137. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having inSIRS or a healthy condition,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group A IRS immune system biomarker genes

as herein defined; and
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group B IRS immune system biomarker genes

as herein defined as herein defined.
138. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having ipSIRS or a healthy condition,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group C IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group D IRS immune system biomarker genes

as herein defined.
139. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having inSIRS or ipSIRS, and wherein
biomarker
values are determined from at least one immune system biomarker in each of
first and
second IRS immune system biomarker groups, wherein:

- 269 -
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group E IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group F IRS immune system biomarker genes

as herein defined.
140. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having inSIRS or ipSIRS, and wherein
biomarker
values are determined from at least one immune system biomarker in each of
first,
second, third and fourth IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group G IRS immune system biomarker genes

as herein defined;
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group H IRS immune system biomarker genes

as herein defined;
c) the third IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group I IRS immune system biomarker genes

as herein defined; and,
d) the fourth IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group J IRS immune system biomarker genes

as herein defined.
141. A method according to claim 131, wherein the first IRS immune system
biomarker is
a PLA2G7 expression product, wherein the second IRS immune system biomarker is
a
PLAC8 expression product, wherein the third IRS immune system biomarker is a
CEACAM4 expression product and wherein the fourth IRS immune system biomarker
is a
LAMP1 expression product.
142. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having mild sepsis or severe sepsis,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:

- 270 -
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group K IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group L IRS immune system biomarker genes

as herein defined.
143. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having mild sepsis or septic shock,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group M IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group N IRS immune system biomarker genes

as herein defined.
144. A method according to any one of the claims 101 to 134, wherein the
indicator is for
determining a likelihood of the subject having severe sepsis or septic shock,
and wherein
biomarker values are determined from at least one immune system biomarker in
each of
first and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group O IRS immune system biomarker genes

as herein defined; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group P IRS immune system biomarker genes

as herein defined.
145. Apparatus for determining an indicator used in assessing a likelihood of
a biological
subject having a presence, absence, degree or prognosis of at least one
medical condition,
the apparatus including a processing device that:
a) determines a plurality of biomarker values, each biomarker value being
indicative of a
value measured or derived for at least one corresponding immune system
biomarker
of the biological subject and being at least partially indicative of a
concentration of
the immune system biomarker in a sample taken from the subject;

- 271 -
b) determines the indicator using a combination of the plurality of biomarker
values,
wherein:
i) at least two biomarkers have a mutual correlation in respect of the at
least one
condition that lies within a mutual correlation range, the mutual correlation
range
being between ~0.9; and,
ii) the indicator has a performance value greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 1 -
BIOMARKER SIGNATURE METHOD, AND APPARATUS AND KITS THEREFOR
[0001] This application claims priority to Australian Provisional Application
No.
2014900363 entitled "Biomarker signature method, and apparatus and kits
therefor" filed 6
February 2014, the entire contents of which are incorporated herein by
reference.
Background of the Invention
[0002] The present invention relates to method, kit and apparatus and to
reagents and
compositions associated therewith for deriving an indicator for use in
diagnosing the
presence, absence or degree of at least one condition in a biological subject
or in prognosing
at least one condition in a biological subject, to a biomarker signature for
use in diagnosing
the presence, absence or degree of at least one condition in a biological
subject or in
prognosing at least one condition in a biological subject, and to a method,
kit and apparatus,
as well as reagents and compositions associated therewith, for identifying
biomarkers for use
in a biomarker signature.
Description of the Prior Art
[0003] The reference in this specification to any prior publication (or
information derived
from it), or to any matter which is known, is not, and should not be taken as
an
acknowledgment or admission or any form of suggestion that the prior
publication (or
information derived from it) or known matter forms part of the common general
knowledge
in the field of endeavor to which this specification relates.
[0004] The analysis of gene expression products for diagnostic purposes is
known. Such
analysis requires identification of one or more genes that can be used to
generate a signature
for use in distinguishing between different conditions. However, such
identification can
require the analysis of many gene expression products, which can be
mathematically
complex, computationally expensive and hence difficult. Much of the biomarker
discovery
process is devoted to identifying a subset of the data that may have relevant
import, from
which a signature is derived using a combination of these values to produce a
model for
diagnostic or prognostic use.
[0005] W02004/044236 describes a method of determining the status of a
subject. In
particular, this is achieved by obtaining subject data including respective
values for each of a
number of parameters, the parameter values being indicative of the current
biological status

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 2 -
of the subject. The subject data are compared to predetermined data that
includes values for
at least some of the parameters and an indication of the condition. The status
of the subject,
and in particular, the presence and/or absence of the one or more conditions,
can then be
determined in accordance with the results of the comparison.
[0006] US2010/0028876 describes methods for diagnosing biological states or
conditions
based on ratios of gene expression data from cell or tissue samples, such as
cancer cell or
tissue samples, by differentiating between cell types, including cancer cell
types. The
invention provides sets of genes that are expressed differentially in normal
and cancer lung
cells and tissues to be able to differentiate these cells and tissues. Such
cellular differentiation
is important in diagnosing cancer and cancer types. The sets of genes are
identified by the
degree (fold change) of up or down regulation. These sets of genes can be used
to
discriminate between normal and malignant cells or tissues, and between
classes of malignant
cells or tissues. Accordingly, diagnostic assays for classification of tumors,
prediction of
tumor outcome, selecting and monitoring treatment regimens and monitoring
tumor
progression/regression also are provided.
[0007] However, traditional methods for biomarker identification and
traditional
combinations of biomarkers use a relatively large number of biomarkers, which
in turn makes
tests expensive to perform, limiting their use in practice. In addition, the
prior art does not
describe the use of immune system biomarker ratios, or a method of identifying
minimal sets
of immune system biomarker ratios useful in determining the presence, absence,
degree or
prognosis of immune system-mediated medical conditions.
Summary of the Present Invention
[0008] In one broad form the present invention seeks to provide a method for
determining an
indicator used in assessing a likelihood of a biological subject having a
presence, absence,
degree or prognosis of at least one medical condition, the method including:
a) determining a pair of biomarker values, each biomarker value being a value
measured or derived for at least one corresponding immune system biomarker of
the biological subject and being at least partially indicative of a
concentration of
the immune system biomarker in a sample taken from the subject;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 3 -
b) determining a derived biomarker value using the pair of biomarker values,
the
derived biomarker value being indicative of a ratio of concentrations of the
pair of
immune system biomarkers; and,
c) determining the indicator using the derived biomarker value.
[0009] Typically the method includes:
a) determining a first derived biomarker value using a first pair of biomarker
values,
the first derived biomarker value being indicative of a ratio of
concentrations of
first and second immune system biomarkers;
b) determining a second derived biomarker value using a second pair of
biomarker
values, the second derived biomarker value being indicative of a ratio of
concentrations of third and fourth immune system biomarkers; and,
c) determining the indicator by combining the first and second derived
biomarker
values.
[0010] Typically the method includes combining the derived biomarker values
using a
combining function, the combining function being at least one of:
a) an additive model;
b) a linear model;
c) a support vector machine;
d) a neural network model;
e) a random forest model;
f) a regression model;
g) a genetic algorithm;
h) an annealing algorithm;
i) a weighted sum;
j) a nearest neighbor model; and,
k) a probabilistic model.
[0011] Typically the method is performed at least in part using an electronic
processing
device.
[0012] Typically the method includes, in at least one electronic processing
device:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 4 -
a) obtaining at least two pairs of measured biomarker values, each measured
biomarker value being a measured value of a corresponding immune system
biomarker of the biological subject;
b) determining a first derived biomarker value indicative of a ratio of
concentrations
of first and second immune system biomarkers;
c) determining a second derived biomarker value indicative of a ratio of third
and
fourth immune system biomarkers; and,
d) determining the indicator by combining the first and second derived
biomarker
values.
[0013] Typically the method includes, in at least one processing device,
generating a
representation of the indicator.
[0014] Typically the representation includes:
a) an alphanumeric indication of the indicator;
b) a graphical indication of a comparison of the indicator to one or more
indicator
references;
c) an alphanumeric indication of a likelihood of the subject having at least
one
medical condition.
[0015] Typically the method includes:
a) comparing the indicator to an indicator reference; and,
b) determining a likelihood in accordance with results of the comparison.
[0016] Typically the indicator reference is based on at least one of:
a) an indicator threshold range;
b) an indicator threshold; and,
c) an indicator distribution.
[0017] Typically the indicator reference is derived from indicators determined
for a number
of individuals in a reference population.
[0018] Typically the indicator reference is based on a distribution of
indicators determined
for a group of a reference population, the group consisting of individuals
diagnosed as having
the medical condition or lacking the medical condition.
[0019] Typically the reference population includes:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 5 -
a) a plurality of individuals of different sexes;
b) a plurality of individuals of different ethnicities;
c) a plurality of healthy individuals;
d) a plurality of individuals suffering from at least one diagnosed medical
condition;
e) a plurality of individuals lacking the at least one diagnosed medical
condition;
f) a plurality of individuals showing clinical signs of at least one
medical condition;
g) first and second groups of individuals, each group of individuals suffering
from a
respective diagnosed medical condition; and,
h) first and second groups of individuals, the first group of individuals
suffering
from a diagnosed medical condition, and the second group lacking the diagnosed

medical condition.
[0020] Typically the indicator is for use in determining the likelihood that a
biological
subject has at least one medical condition, and wherein the reference
population includes:
a) individuals presenting with clinical signs of the at least one medical
condition;
b) individuals diagnosed as having the at least one medical condition;
c) individuals diagnosed as lacking the at least one medical condition; and,
d) healthy individuals.
[0021] Typically the indicator reference is retrieved from a database.
[0022] Typically the likelihood is based on a probability generated using the
results of the
comparison.
[0023] Typically the indicator is for determining a likelihood of the subject
having a first or
second condition, and wherein the method includes:
a) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of first and second conditions;
and,
b) determining the likelihood in accordance with the results of the
comparison.
[0024] Typically the method includes:
a) determining first and second indicator probabilities using the results of
the
comparisons; and,
b) combining the first and second indicator probabilities to determine a
condition
probability indicative of the likelihood.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-6-
100251 Typically the first and second indicator references are distributions
of indicators
determined for first and second groups of a reference population, the first
and second group
consisting of individuals diagnosed with the first or second condition
respectively.
[0026] Typically the method includes:
a) obtaining a sample taken from the biological subject, the sample including
polynucleotide expression products; and,
b) quantifying at least some of the polynucleotide expression products within
the
sample to determine the pair of biomarker values.
[0027] Typically the method includes, determining the indicator at least in
part using a ratio
of concentrations of the polynucleotide expression products.
[0028] Typically the method includes:
a) quantifying polynucleotide expression products by:
b) amplifying at least some polynucleotide expression products in the sample;
and,
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products; and,
d) determining the indicator by determining a difference between the
amplification
amounts.
[0029] Typically the amplification amount is at least one of:
a) a cycle time;
b) a number of cycles;
c) a cycle threshold;
d) an amplification time; and,
e) relative to an amplification amount of another amplified product.
[0030] Typically the method includes determining:
a) a first derived biomarker value by determining a difference between the
amplification amounts of a first pair of polynucleotide expression products;
b) a second derived biomarker value by determining a difference between the
amplification amounts of a second pair of polynucleotide expression products;
c) determining the indicator by adding the first and second derived biomarker
values.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-7-
100311 Typically the immune system biomarker is a biomarker of an immune
system of the
biological subject that is altered, or whose level of expression is altered,
as part of an
inflammatory response to damage or pathogenic insult.
[0032] Typically:
a) the at least two immune system biomarkers have a mutual correlation in
respect of
the at least one condition that lies within a mutual correlation range, the
mutual
correlation range being between 0.9; and,
b) the indicator has a performance value greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.
[0033] Typically the mutual correlation range is at least one of:
a) 0.8;
b) 0.7;
c) 0.6;
d) 0.5;
e) 0.4;
f) 0.3;
g) 0.2; and,
h) 0.1.
[0034] Typically each immune system biomarker has a condition correlation with
the
presence, absence, degree or prognosis of the at least one condition that lies
outside a
condition correlation range, the condition correlation range being between
0.3.
[0035] Typically the condition correlation range is at least one of:
a) 0.9;
b) 0.8;
c) 0.7;
d) 0.6;
e) 0.5; and,
f) 0.4.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-8-
100361 Typically the performance threshold is indicative of an explained
variance of at least
one of:
a) 0.4;
b) 0.5;
c) 0.6;
d) 0.7;
e) 0.8; and,
f) 0.9.
[0037] Typically the immune system biomarker value is indicative of a level or
abundance of
a molecule selected from one or more of a nucleic acid molecule and a
proteinaceous
molecule.
[0038] In some embodiments, the indicator is for determining a likelihood of
the subject
having inSIRS or ipSIRS, and wherein the method includes:
a) determining a first pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the PLA2G7 gene and PLAC8 gene;
b) determining a second pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the CEACAM4 gene and LAMP] gene; and,
c) determining the indicator using the first and second pairs of biomarker
values.
[0039] In some embodiments, the indicator is for determining a likelihood of
the subject
having inSIRS or a healthy condition, and wherein biomarker values are
determined from at
least one inflammatory response syndrome (IRS) immune system biomarker in each
of first
and second IRS immune system biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: NUMB, RAB27A, USP3//LOC100130855, HIF1A,
LBXCOR1//PIAS1//CALML4, SQRDL, C20orf74, ILlORB, PARP4, DNTTIP1,
MTMR6//L0C646482, LAMP2, MAPK1, SERINC3//TTPAL, IGSF6//METTL9,
RP2, Cl8orf32, L0C284757, MTMR10//MTMR15, SLC12A6, LCP1, CHP,
PRR13, C20orf177, ZFP106, DICER], PHF12, IFNAR1, BNIP2, UBE2A, NIN,
MBD2//SNORA37, TM9SF2, RAB8B, CLIP], WAS, DNAJC3, CDADC1,
KIAA0317, MED13L, INTS6, PDK3, MY05A, NUPL1, VEZF 1, CUL4B,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 9 -
USP9X//USP9Y, RPS6KA3, IL17RA//CECR7, ELF], TMX4, TA OK], ELM02,
STAT5B//STAT5A,
PAN3//EEF1A1//CHCHD2,
SIPA1L1//SNORD56B//LOC145474//LOC283567, OSBPL1A, SYNI1, U2AF1,
NPEPPS//TBC1D3F//L0C440434, AP1G1, SNTB2, ZNF230//ZNF222,
ME2, GALNT1, DYRK1A, ZMYM2, ARID4A, TOB1, DOCK]], ACTR10,
ZMYM5//ZMYM2, FNDC3A, NUFIP2, STRADA, SPG11//ISLR,
SPATA13//C1QTNF9, BRWD3, BACH1, CLTC, LIG4, C2 lorf41//BACH1,
KPNB1, DHRS7, USP8, LACTB, SYNE2, ZDHHC20//LOC728099, EAPP,
MED13//LOC100129112, TAOK3, NLGN3, CIT, RIPK3, CP110, ABHD2,
GNA13, GGNBP2, PXN and PTPN1 (hereafter referred to interchangeably herein
as "group A IRS immune system biomarker genes" or "group A IRS biomarker
genes"); and
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: AGFG1, BMX//HNRPDL, MCM5, TRIM28, GRWD1, ZNF574,
ARRDC2, PELP1, SHPK, GPS1, FAM38A, FBX031, Cl6orf58//LOC100128371,
NLRC3, JMID8, CDK10, TRAPPC2L, PRI1T7, BRF1,
MTA1//LOC647310//LOC100128343, PLD4, DDX54//CCDC42B, PLBD1,
IRAK3, FGD4, ARG1, RANGAP1, UNC84B, SAMSN1//L0C388813, PFKL,
S100Al2, KIF22, LRRN1, CCDC134, LZTR1, GZW, ICAM2, TMC8,
LAT//SPNS1//NPIPL2//LOC728741//LOC730153//NPIPL3//SPIN1//LOC728888//
LOCI 00289169//LOC728734//LOC729602//LOC100288442//LOC100288332,
CLEC4D, CDK5RAP1, PPP1R16B, DAZAP1, LMF1, EDC4, IL21R//L0C283888,
JMID7-PLA2G4B//JMID7//PLA2G4B,
TMEA1120B//RHOF,
ENTPD1//C 1 Oorf131, ACSL1, ZC3H7B, CHERP//C19orf44//CALR3, U2AF2,
PYGL, SOS2, ANKRD22, MEGF9, MGAM//LOC100124692, IL1R2, IL2RB,
FCAR, IL27RA, DHX37, PATZ1, PRDM15, NOSIP, RPTOR, SPG7, DNAJA3,
127\TN1, SEPT9, THAP11, LPCAT2, MAN2C1, PITPNM2,
NOC2L//SAMD11//LOC401010, ZMYM3,
KCNE1, ACTR5, FAM110A,
FAM134C, LLGL2, INF2, KDM2B, ACSL4, B4GALT5, CD79B, BCL11B,
ERLIN1, TLR4, EVL, SRGN, 5LC37A3, GPR141, MSL3, WP9//LOC100128028,
MAP2K6, PHTF1, KLHL36, POLR3E, PCNX, PNKP, TMEM104, TRPV2,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 10 -
SEPT], APH 1B, POLE, MED24, MPI, C 1 2orf49, PES1, ERCC1//CD3EAP,
CD177, CPD, MEF2A//LYSMD4, C 1 4orf43 , RPLPO, CDC25B, SYMPK,
ARHGEF 18//LOC 100128573,
PSTPIP2,
HERC2//HERC2P2//HERC2P3//L0C440248, MAPK14, F5, PLCG1, ZNF416,
AARS, KLHL2, APOBEC3A//APOBEC3B, CMTM1//CKLF, USP 11,
M4P3K14//LOC 100133991, GOLGA3, TMEA1204, S100A8, IL1R1, DHPS,
PPP2R1A, UBTF, DRG2, DNMT1, U5P36, ZBTB4, TSC2, KI4A0195, KIAA0182,
ALOX5AP, TGIF2, 5T20//C15orf37, FN3KRP, ABCD4, ZFP64, NE01,
PPIL2//YPEL 1, RNPS1, NF2, SERPINB 1, DDX51, PRPF6, TIA4M22,
SYS1//SYS1-DBNDD2//DBNDD2, RAB31, KRH , SMARCA4, CLUAP 1, C 1 6orf67,
C20orf4, CHTF8//HAS3, NPTN, CSRP2BP, AES, ODZ 1, MTMR15//MTMR10,
SIRPD, EEF2//SNORD37, DKCP/SNORA36A//SNORA56, CEACAM4, C 1 2orf43,
RANBP3, EEF2K, L0C338799, PLP2, AKAP8, ELAC2, AKAP 1, TBC 1D4,
ALOX5, WSB 1, BAZ 1A, ETS2, GGA2, CSTF2//RAD21, METTL9, GYG1,
CRAMP 1L//HN1L, EVI2B, PPP 1R13B, POPS, C20orf3, WDR59, KCNI15,
PGLYRP 1, ELAVL1, SLC25A 1, PSMD3, CDC42EP3, FTSj3, C2CD2, RBM19,
CDH26, TRA1T2B,
GTF2F1//LOC 100130856,
SNRPN//SNURF//IPW//SNORD 116-16//SNORD 116-18//SNORD 116-
21//SNORD 116-22//SNORD 116-171/SNORD 116-19//PAR5//PAR-
SN//SNORD 116-2//SNORD 116-25//SNORD 116-26//SNORD 107//SNORD 115-
12//SNORD 115-5//SNORD 115-6//SNORD 115-9//SNORD 116-11//SNORD 116-
12//SNORD 116-13//SNORD 116-28//SNORD 116-
4//SNORD64//PAR1//SNORD 109A//SNORD 109B//SNORD 116-6//SNORD 116-
3//SNORD 116-9//SNORD 115-13//SNORD 115-1//SNORD 115-14//SNORD 115-
15//SNORD 115-21//SNORD 115-10//SNORD 115-7//SNORD 115-16//SNORD 115-
40//SNORD 115-42//SNORD 115-11//SNORD 115-29//SNORD 115-34//SNORD 115-
36//SNORD 115-4//SNORD 115-43//HBII-52-24//SNORD 116-5//SNORD 116-
7//SNORD 115-26//SNORD 115-30 SNORD 116-15//SNORD 116-8//SNORD 115-
2//SNORD 115-39//SNORD 116-14 SNORD 116-20//SNORD 115-8//SNORD 115-
3//SNORD 115-38//SNORD 115-41 SNORD 115-22//SNORD 115-44//SNORD 116-
1//SNORD 115-17//SNORD 115-18//SNORD 115-19//SNORD 115-
20//SNORD 116@, SLC9A8, RPA1, ADARB1, AFG3L2, MCTP2, DACH1, SEH1L,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 11 -
RRP 1B, ZNF335, WDR73, TAF15, MOSPD2, WIPI1//ARSG, ARRB2,
PLIN5//LRG1, SNRPD3//C22orf13, CTNNBL1, ZNF 175, NCF4, DDX27,
FBX021, TDP 1, ATXN2L, ILF3, VAPA, DDX19B//DDX19A, NCOR2, KL,
MTHFS, TOM1L2//L0C246315, APOBEC3D,
EXD2,
CDR2//RRN3//LOC100131998//L0C653390, ADCY4, DHX33, CKLF, GTF3C1,
PRKCSH, DHX35, HSPH1, CCDC92, BCOR, CCPG1//PIGB//DYX1C1,
MCM3AP, FPR1, ZNF460, AKAP8L, DCAF7, RNF24, NSMCE1, PDHAl,
SAFB2//SAFB, ITM2B, ZNF236, PI4KA//PI4KAP1//PI4KAP2//LOC100293141,
CSTB, C14orf138, ITGAL, ARID3A, COG 7, TYROBP, HP//HPR,
SRCAP//SNORA30, COG], GK//GK3P//FTL//L0C652904, C15orf63//SERF2,
SERPINA6, SMG6//C17orf6, IN080, C16orf62, RAB35, PEF 1, Cl4orf101,
TMEM185A, LIMK2, CTCF, DIABLO//B3GNT4, VPS33A, UNQ1887,
TBCB//POLR2I, ABHD13, SLC24A6, EDNRB, CA12, ANAPC5, TMC3, TRIAP 1,
ABHD 12B, TDRD9, EIF2B 1,
CXorf59,
LRRC37A3//LRRC37A2//LRRC37A//ARL 17P 1//LRRC37A4//LOC 1 00294335//L0
C644397, SDS, SYCP2, TBC1D8B, TMEM31, GUCY 1B2, PFN1, SLC24A3,
ABCC11//LONP2 and ZNF257//ZNF492//ZNF99//ZNF98//L0C646864 (hereafter
referred to interchangeably herein as "group B IRS immune system biomarker
genes" or "group B IRS biomarker genes").
[0040] In some embodiments, the indicator is for determining a likelihood of
the subject
having ipSIRS or a healthy condition, and wherein biomarker values are
determined from at
least one IRS immune system biomarker in each of first and second IRS immune
system
biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: LTBP3, LPHN1, NR1D1//THRA, METTL9, PLD4, MAPK14,
FAM102A, MYBBP1A//SPNS2, FLJ10232, SEMA4C, LMF1, PLBD1, MAN1C1,
B4GALT5, ENGASE, NDRG2, TLR5, WDR4, PATZ1, CD177, LILRA5, SIRPD,
ADAMTS10, TCF7, GGT7, GYG1, CDAN1, BRF1, GPR84,
TMC6//LOC100131096, PGAP3,
GRAP//SNORD3B-1//SNORD3B-
2//L0C400581, SHPK, NOG, PVRIG//PILRB//STAG3, TDRD9 , TMC8, C 1 4orf21,
NLRC4, APBA2, TBL3, LDOC1L, C16orf58//L0C100128371, KLHL3, IRAK3,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 12 -
JMJD7-PLA2G4B//JMJD7//PLA2G4B, RAB31,
ILlORB,
NFATC1//LOC100127994, S100Al2, SLC2A3//SLC2A14, GPA33, PLXDC1,
BCL6//LOC100131635, GTPBP3, SQRDL, CD7, TMEM177//L0C100125918,
FBX031, CACNA1E, LILRA4, FAM38A, UBE2J1, SBF1//SBF1P1, TMEA1204,
FCAR, CDK4/MARCH9/C3HC4, ZBTB4, PIWIL4, RNASE2//L0C643332,
PRA1T7, AGFG1, CMTM1//CKLF, MCTP1, ARL11, FMNL3//PRPF40B,
FAM151B, BAHD1, OSBPL7, ZAP70, TUBGCP6, MAP2K6, NPTN, C3AR1,
CD247, S100A8, TBCD, LMNB1//PCIF1, PFKL, GRWD1, PYGL, UPP1, OMG,
SAMSN1//L0C388813, BLCAP, PTPRS, FAM20A, CARD6, SPPL2B, IL2RB,
SORT], BST], TAF1C//ADAD2, SEA/L614F, NCAPH2, MCTP2, ZAK, CCR7,
MAN2C1, NEURL4//GPS2//D45234E, BMX//HNRPDL, TRAPPC6A, LPCAT2,
Cl9orf60, SLC4A10, Cl4orf101, TP53I3, IL1RN, AIM2, UBE2R2, PNKP,
ZNF70, SEPT], NE01, MPRIP, DPH1//0VCA2, Cl6orf67, CD58, RAB27A,
EEF2K, CLIC1, MBLAC2, IFNGR2, CRTC1//M4ML2, CACNB1, GALNT3,
C 19orf6, C20orf74, RALB, GPRASP1, CA4, ETS2, RP2, MARS2, RAB32, FAIM3,
C20orf24//SLA2, ZNF549, PIGL, PHTF1, IL18R1, IP04, ZFP106, SLC12A4,
DNTTIP1, S100A11, ZNF544, ATXN1, GNLY, MID2, BACH2, INF2, ARFGAP1,
MSL3, SOS2, ARL8A, PTPRKOR4B1, NAT9, RHOT2//FBXL16, PNPLA1,
DNAJC13, GNG5, FAM129C, PXK, ClOorf119, BATF, LM07, KLF2, NRD1,
CLCN7, GLA, CFLAR, SYCP2, IMAGE5303689, LPGAT1, PTGDR, LAMP2,
ZNF607, INSL3//JAK3, DUSP3, PCNX, CD79B, IRAK], ZNF550//ZNF549,
LOC100130950, SPTLC2, CTSA, RAP2C, ADCY9, MED12L, MTHFD2, CAP],
TOR1AIP2//TOR1AIP1//IFRG15, CHP, TSEN2, LYRAll, UBE2A, NUPL1,
YIPF1, FRA1D3, KAL1, CLTC, FLVCR2//RP524, WSB2, KIAA0040, JAG],
GPR183, N6AMT1, ZNF563, AP3B2, SERPINB2//SERPINB10, CDH2, ITFG1,
EDEA12, RNF135, HPSE, DSC1, FOXN2, RASSF2, ZNF420, ZFP28,
UBE2G2//SUM03, PTTGlIP, PRKCB, KIT, PLEK, MAP4K4, GBA//GBAP,
CLIP], EDEM3, SERINC3//TTPAL, TPST2, HNRPLL, TP53BP2, KCND1,
GCLM, RIT1, OSCAR, DDX59, EDNRB, ELM02, RRAGC, AFTPH,
DCUN1D3//LYRA11, RSBN1, IFI30, SNX1, PTPN1, SEP15, ARA1CX5, ALAS],
NFKBIA, STXBP2//L0C554363//LOC100131801, Cl5orf24, SRI, ASGR2,
NSF//L0C728806, TRIM69, SEC23A,
PLAUR,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 13 -
RAB3GAP2//AURKAPS1//AURKA//SNORA36B, M40B//NAT13, DEDD,
SEC23B, COPA, EGF, STRADA, SIAE, C5, SLC30A1, ANXA4, NKG7,
ABHD12B, TESK2, LONRF3, PIKFYVE, SH3BGRL3, ARIICX3, NEU1, SPAST,
STX6//KI4A1614, TADA3L, LIN37//PSENEN, UBR3, WDR90, RTN2 and TMUB2
(hereafter referred interchangeably herein as "group C IRS immune system
biomarker genes" or "group C IRS biomarker genes"); and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: CNNM3, SLC25A45, UNC84B, ARHGEF18//LOC100128573,
PIK3IP1, EPHX2, SEPT9, ITPKB, TSPYL2//GPR173, GALT, USP11,
CBX7//LOC100128400, MAP3K14//LOC100133991, CDK5RAP 1, KLHL36,
SPG7, ZNF574, RASA3, KLHL22//KRT18, PYG02, U5P36, LCK, SKI,
C5orf45//SQS1M1, PIK3C2B//L0C100130573, ANGEL], ZCCHC14, CIRBP,
ZMIZ2, TMFM120B//RHOF, NOC2L//SAMD11//LOC401010, PPP1R13B,
ZNF416, PBXIP1, SMYD5//NOTO, ZNF529, EDC4, LENG8,
TBC1D22A//L0C100289878, COR07, COG8//PDF, CUL9, RASGRF2,
CHERP//C19orf44//CALR3, POLR3E, CNNM4, TSC2, XYLT2//L0C100130580,
TP53, LMBR1L, AKT1, SLC7A6//SLC7A60S, LDLRAP1, SGSM2,
ZNF 764//ZNF 747 , AKAP 1, RNPS 1 , ICAM2 , KIF3A,
TGIF2,
VAC14//L0C100130894, CXXC5, DCAF15, TARBP1, RCAN3, IMP4, LUC7L,
SIN3B, TRI1T2B, POFUT2, AXIN1, PPIL2//YPEL1, NLRP1//L0C728392,
TMEA163A, TMEM208, ZNF362, GNG7, CASS4, ZNF287, DGKE, CEP68,
ASXL1, CLUAP1, WDR89, CD4OLG, MGC57346//C17orf69, PPIE//CCDC25,
FCH01, TNP02//SNORD41, CSNK1E//L0C400927, CLYBL, XAB2,
METT1 OD//LOC 284009 , PRPF6, AKAP8, SH3BP
1//PDXP,
TOM1L2//L0C246315, ZNF329, ZNF274, FAM119A, SMYD3,
UNC84A//C7orf20, ZNF256, PSD4, CCDC130, LIMD2//MAP3K3, ELP2, ZNF8,
AFG3L2, TXK, DDX27, FBXL12, TNRC6C, TADA1L, KIAA0355//FLI21369,
ZNF211, ZNF808//ZNF578//ZNF611, SRCAP//SNORA30, BANP//RUNDC2C,
ADARB1, CCDC71, KTI12, TCF25, XYLT1//LYR112//ZC3H11A, DET1, ABCF3,
PRKCZ, KIAA0141, CHI3L1, RPGRIP1, TTC31, MTMR15//MTMR10, MEF2D,
TMEM50B, GLOD4, PRPF8, C14orf43, P2RX5, MSH2, PCCA, DENND4B,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 14 -
SLC43A2, MAPK8IP3, TUBGCP5, C19orf2, SEH1L, CCDC104, TRIM62,
TDRKH, COG], POLR1B, AFG3L 1, TYK2, RBM3, UBTF, RP 11-
9412.2//NBPF16//NBPF11//NBPF15//NBPF8//NBPF20//NBPF10//NBPF14//NBP
F1//LOC100288142//NBPF12//KIAA1245//LOC100290137, ZNF41, ZNF461,
PI4KA//P14KAP 1//PI4KAP2//LOC 1 00293141, THEA14, BCL 11A, CC2D 1B,
WDR7 3, BBS2//0GFOD 1, RRN3//L0C653390//L0C730092//LOC100131998,
N0P58, NUCKS1, ZNHIT6, RXRB, AKT3, FANCM, ERN], FAM117B,
COX11//TOM1L1, ACVR2A, RP3-402G11.5, AHCTF 1, CLN8, NVL, SAPS2,
DPEP3, PDE3B, DPEP2, GGA1, CCDC50, SNRPN//SNURF//IPW//SNORD116-
16//SNORD116-18//SNORD116-21//SNORD116-22//SNORD116-17//SNORD116-
19//PAR5//PAR-SN//SNORD116-2//SNORD116-2.5//SNORD116-
26//SNORD107//SNORD115-121/SNORD115-5//SNORD115-6//SNORD 115-
9//SNORD116-11//SNORD116-12//SNORD116-13//SNORD116-28//SNORD116-
4//SNORD64//PAR1//SNORD 109A//SNORD 109B//SNORD116-6//SNORD116-
3//SNORD 116-9//SNORD115-13//SNORD115-1//SNORD115-14//SNORD115-
15//SNORD115-21//SNORD115-10//SNORD 115-7//SNORD115-16//SNORD115-
40//SNORD 115-42//SNORD 115-11//SNORD 115-29//SNORD115-34//SNORD115-
36//SNORD115-4//SNORD115-43//HBI1-52-24//SNORD116-5//SNORD116-
7//SNORD 115-26//SNORD 115-30 "SNORD 116-15//SNORD116-8//SNORD115-
2//SNORD115-39//SNORD116-14 SNORD116-20//SNORD115-8//SNORD115-
3//SNORD115-38//SNORD115-41 SNORD115-22//SNORD115-44//SNORD116-
1//SNORD 115-17//SNORD 115-18//SNORD 115-19//SNORD 115-
20//SNORD116@, C 17orf65//ASB16, ZNF317, SNRNP200, CXorf26, MTBP,
NOL11//SNORA38B, CCNL2, ALDOC, PITPNC1, FASTKD2, ZZZ3, PIK3R5,
WDR82, GLDN, CHML, C 1 5orf40, DID01, CLCC1//GPSM2//C 1 orf62 ,
SLC35D1, SCRN1, C 1 5orf63//SERF2, ZNF460, SAFB2//SAFB, C] 6orf54,
DDX18, CTPS2, ZNF382, ZNF101, LIPT1//MRPL30, ITGA6, KIF21B, INPP5B,
SF3A1//CCDC157, ODF2L, NUAK2, CHCHD5, AHSA21/U5P 34, YLPM1,
TERF2, ZNF830, MAN2B1//MORG1, GPATCH8, SHC1, SEPT4, SFRS2, TMC3,
OTUD5, NARG1L, MKL1//KIAA1659, YTHDF1, SLC14A2, GGA3 and EXOC7
(hereafter referred to interchangeably herein as "group D IRS immune system
biomarker genes" or "group D IRS biomarker genes").

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 15 -
[0041] In some embodiments, the indicator is for determining a likelihood of
the subject
having inSIRS or ipSIRS, and wherein biomarker values are determined from at
least one
IRS immune system biomarker in each of first and second IRS immune system
biomarker
groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: ALKBH5 // FLI13773, RPS19BP1, RFXANK // MEF2B //
L00729991, NA, CDC6, Cl9orf56, NA, ABCC2, THAP11, RTN2, MAZ,
TAX1BP3, NUTF2, MPZL3, FBXW5, HIST1H2BM, CETP, PQLC1, H2AFX,
KIAA0101 // CSNK1G1, STK17B, SMARCD3, L0C100134934 // CDK3,
LPCAT4, LPP, MPZL2, ANKRD9, PRR13 // PCBP2, MDS2, RBM33, GATAD2B
// PLIN2, PPTC7, MYBL2, 01P5, PLA2G7, CRIPT, RNF186, CCDC125, TLE3,
C3orf35, SAP130, MXD1, ZHX2, CDK5RAP3, ENTPD1 // ClOorf131, NDUFB7,
POGZ, DOK3, MCM7, IL17F, CLPS and DUSP11 (hereafter referred to
interchangeably herein as "group E IRS immune system biomarker genes" or
"group E IRS biomarker genes"); and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: PIWIL4, Cl lorf82, ACRC, CLP7M1L, MAGED2, PLAC8,
ZDHHC4, OTX1, INSIG1, BATF, MFSD11, DNASE1L1 // RPL10, Cl5orf24,
CDS2, KEAP1, ARD1A, POLR2G, AHCY, SLC39A9, CUGBP1, FAM96B,
TM7SF3, CTSZ, CD63, SPPL2A, ST3GAL2, SEC13, TM9SF1, IRAK2, GOSR2,
ADIPOR2, TG, GABRR2, TPST2, DERL2, CCDC101 // L0C388242, VWA5A //
OR10D1P, CD300A, MRPS34, PSMA7, MAPK6, JKAMP, TLR10, RAG1AP1,
NEU1, SLC30A1, PDGFC, ATOX1, CYBASC3, TMFM205 // hCG 29977,
FAM108B1, ACSS2, HIST1H4L, AGTRAP, RNF114, UBQLN2, EDF1,
C20orf197, UBE2E1, RER1, ANKRD10, SEC22C, TM2D3, SLC15A2, TRIM28,
COX15, CCDC109A, CSTF1, AIP, ACTR1A, HIST1H4I, YIF1B, TSPAN31,
VPS26B, CNIH, TGFBR1, NIPA2 // CYFIP1, DDAH2, BID, CYB5R1, CEACAM4,
KIT, GAB2, JAG_ RPGRIP1, VAT], GNB5 // LOC100129973, SSR4 // IDH3G,
LAMP], MRPL41, RUNX2, ITFG1, DNA SE], ZNRD1 // NCRNA00171, NLRP1 //
L00728392, STT3A, MGAT4B // SQS7M1, KIAA1257 // ACAD9 //

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 16 -
LOC100132731, KLHL6, PTPN6, GALK2, DAD] // OR6J1, PDLIM5, TMEM-147,
TRAM] // L0C286190, LZTR1, TNP01, ACSL5, C22orf37, PLK1, SYNE2,
PSMD3, FLJ27255, PRKCD, RAB34, RPN2 // EEF1A2, SLC35B1, KCNIP3,
PDE3B, PXMP2 // PGAM5, SDF2, HIST1H3I, L0C284757, TMEA133 //
DCAF4L1, CSNK2A2, LSM10, PTTGlIP, ADRB3 // GOT1L1, PLXNA2, DIAPH2,
BICD2, HAL, RPS6KC1, TMEM106C, CD1E, SLC35A5, C7orf26, IMP3,
PICALM, ARF1, FHOD1 // SLC9A5, Cl9orf55, TOW40L // NR1I3, INSIG2,
NEK9, HCG27, SDHB, CUBN, PRDX3, CEPT1 // DRAM2, ERGIC1, KPNA3,
VAV1, ELM01, CUGBP2, LASP1, COL9A2, MEGF9, ELF4, SUZ12P, SULT1A2
// SULT1A1, FAM123C, FAR2, IER2, RGS2, MYBPH, MFAP3, RCHY1, MGAT1,
MFSD4, CDH2, TMEM184C, CTRB2 // CTRB1,MPP4, PHF12, SLBP, ADAM19,
HTR1B, TRIM55, CRNN, KLHDC7A, YIPF5, SLC11A1, GABBR1, CAMKV,
SLC35F5, CHRNG, CXCL14, METTL6, PHC2, GPR153, TNFRSF10D, BAT2L,
GALNT2, DENND3 // C8orf60, CLDN3, F11, CCDC93, FLJ46365, CYP21A2,
ETV5, TRPM2, IL20, NBL1 // Clorf151, NGEF, POU6F2, PTEN // PTENP1,
NPC1L1, CYP4B1, NFIC, PPARGC1A, PLIN3, THPO, TIMP4, CELSR2,
DMBX1, CAMK2B, PPFIA1, HCLS1, SLC6A20, Cl7orf66 // RSL24D1, PIWIL2,
DAZL // DAZ4 // DAZ3 // DAZ2, GAL3ST2, TPD52L1, Cl9orf34, RASGEF1C,
BAALC // F1,110489, NR4A2 // FLJ46875, HAPLN1, CLDN18, TAS1R1, TIMD4,
SKI, CCDC48, MEGF10, OSBPL6, DNAH2, ARID1B, PGC, DCST1, SDK],
CHIA, REG4 // NBPF7, TMEA149 // CLTC // MIR21, TMEA144, NDUFB6 //
DFFB, COL25A1, EPHX4, NCRNA00085, NTRK3, PKHD1, SLC2A7, NTRK1,
ABHD1 // PREB, SLC4A9, GPNMB, SLC5A1, SLC7A8, RTCD1, PROC,
Cl7orf64, FL114100 // Clorf86, NUP50, UNKL, ClOorf18, TMEM61, C9orf68,
CYTSA, MORN3, RAB17, CNNM3, CCDC28B, SH2D6, BARHL2, T, SNRK,
TCP11, KDR, ENA114, UNKL, SPRR2C, GPR17 // LOC100291428 // LIMS2,
Clorf175 // TTC4, CACNA1D, C2orf62, LOC100132686, UNQ6126, TRIM15,
GPR113 // SELI, IL22, SCN10A, FAAH,MBOAT7, C7orf51, KI4A1530, TRPM8,
Clorf95, DDC // LOC100129427, GABRA1, HCRTR1, DCST2, CHODL, PAN3 //
EEF1A1 // CHCHD2, LYPD6B, UGT3A1 and SERPINA6 (hereafter referred to
interchangeably herein as "group F IRS immune system biomarker genes" or
"group F IRS biomarker genes").

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 17 -
[0042] In some embodiments, the indicator is for determining a likelihood of
the subject
having inSIRS or ipSIRS, and wherein biomarker values are determined from at
least one
IRS immune system biomarker in each of first, second, third and fourth IRS
immune system
biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: RPS19BP1, RFXANK//MEF2B//LOC729991, C19orf56, RTN2,
HIST1H2BM, CETP, PQLC1, H2AFX, KIAA0101//CSNK1G1,
L0C100134934//CDK3, LPCAT4, LPP, MPZL2, ANKRD9, RBM33, MYBL2,
PLA2G7, 01P5, CRIPT, RNF186, C3orf35, ZHX2, NDUFB7 and DUSP11
(hereafter referred to interchangeably herein as "group G IRS immune system
biomarker genes" or "group G IRS biomarker genes");
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: PIWIL4, C11orf82, ACRC, PLAC8, ZDHHC4, OTX1, INSIG1,
BATF, MFSD11, C15orf24, CDS2, POLR2G, 5LC39A9, FAM96B, TM7SF3,
SPPL2A, ADIPOR2, GOSR2, DERL2, TPST2, VWA5A//0R10D1P,
CCDC101//L0C388242, MAPK6, PSMA7, JKAMP, TLR10, RAG1AP1,
SLC30A1, PDGFC, ATOX1, TMEM205//hCG 29977, FAM108B1, UBQLN2,
EDF1, C20orf197, RER1, UBE2E1, ANKRD10, SEC22C, TM2D3, SLC15A2,
CCDC109A, HIST1H4I, TSPAN31, TGFBR1, CNIH, DDAH2, NIPA2//CYFIP1,
BID, CYB5R1, KIT, RPGRIP1, MRPL41, RUNX2, ITFG1,
ZNRD1//NCRNA00171, NLRP 1//LOC728392,
KIAA1257//ACAD9//LOC100132731, KLHL6, GALK2, DAD1//0R6J1, PDLIM5,
TRAM1//LOC286190, 17\TP01, ACSL5, SYNE2, RPN2//EEF1A2, SLC35B1,
KCNIP3, TMEM33//DCAF4L1, CSNK2A2, LSM10, PLXNA2, DIAPH2, HAL,
RPS6KC1, 5LC35A5, PICALM, C19orf55, INSIG2, SDHB, PRDX3,
CEPT1//DRAM2, KPNA3, SULT1A2//SULT1A1, FAR2, MYBPH, MFAP3,
RCHY1, CDH2, TMEM184C, CTRB2//CTRB1, SLBP, CRNN, YIPF5, CHRNG,
5LC35F5, METTL6, CLDN3, CCDC93, CYP21A2, NBL1//Clorf151, NGEF,
POU6F2, NPC1L1, PPARGC1A, THPO, CELSR2, DMBX1, SLC6A20,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 18 -
Cl 7orf66//RSL24D1, GAL3ST2, C19orf34, BAALC//FLJ10489, CLDN18,
TAS1R1, CCDC48, OSBPL6, SDK1, TMEA149//CLTC//MIR21, TMEA144,
NDUFB6//DFFB, NCRNA00085, NTRK3, NTRK1, SLC4A9, SLC5A1, RTCD1,
FLI14100//Clorf86, PROC, Cl 7orf64, UNKL, C9orf68, MORN3, RAB17,
CNNM3, CCDC28B, BARHL2, UNKL, L0C100132686, UNQ6126, IL22, FAAH,
C7orf51, DCST2, LYPD6B and SERPINA6 (hereafter referred to interchangeably
herein as "group H IRS immune system biomarker genes" or "group H IRS
biomarker genes");
c) the third IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: ALKBH5//FL113773, RNA243, ARD1A, CDC6, AHCY,
MRPS34, CYBASC3, HIST1H4L, RNF114, TRIM28, CSTF1, CEACAM4, GAB2,
GNB5//L0C100129973, THAP11, SSR4//IDH3G, STT3A, NUTF2, MPZL3,
TMEM147, RAB34, PDE3B, PXMP2//PGAM5, HIST1H3I, L0C284757,
TMEM-106C, STK17B, IMP3, HCG27, CUBN, ERGIC1, ELMO], CUGBP2,
COL9A2, MEGF9, SUZ12P, FAM123C, RGS2, PRR13//PCBP2, PHF12,
ADAM19, GATAD2B//PLIN2, SLC11A1, PPTC7, PHC2, BAT2L,
DENND3//C8orf60, F1.146365, ETV5, CCDC125, PTEN//PTENP1, TLE3, NFIC,
TIMP4, PPFIA1, HCLS1, SAP130, MXD1, NR4A2//FLJ46875, SKI, ARID1B,
ENTPD1//ClOorf131, POGZ, DOK3, REG4//NBPF7, MCM7, SLC7A8, NUP50,
ClOorf18, TMEM61, 5H2D6, SNRK, SPRR2C, CACNA1D, TRIM15, CLPS,
MBOAT7, KIAA1530, C1orf95, GA BRA], HCRTR1, CHODL and
PAN3//EEF1A1//CHCHD2 (hereafter referred to interchangeably herein as "group
I IRS immune system biomarker genes" or "group I IRS biomarker genes"); and,
d) the fourth IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: CLP1M1L, MAGED2, DNASE1L1//RPL10, KEAP1, CUGBP1,
CTSZ, CD63, ST3GAL2, SEC13, TM9SF1, IRAK2, GABRR2, TG, CD300A,
NEU1, ACSS2, AGTRAP, UPF0627, COX15, ABCC2, ACTR1A, AIP, YIF1B,
VPS26B, JAG], VAT], LAMP], DNA SE], MAZ, TAX1BP3, MGAT4B//SQS1M1,
FBXW5, PTPN6, LZTR1, PLK1, C22orf37, PSMD3, FLJ27255, PRKCD, SDF2,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 19 -
PTTGlIP, ADRB3//GOT1L1, BICD2, CD1E, C7orf26, ARF1, FHOD1//SLC9A5,
TOW4OL//NR1I3, NEK9, VA Vi, SMARCD3, LASP1, ELF4, IER2, MGAT1,
MFSD4, MDS2, MPP4, HTR1B, TRIM55, KLHDC7A, GABBR1, CAMKV,
CXCL14, GPR153, TNFRSF10D, GALNT2, F11, IL20, TRPM2, CYP4B1, PLIN3,
CAMK2B, PIWIL2, DAZL//DAZ4//DAZ3//DAZ2, TPD52L1, RASGEF1C,
HAPLN1, TIMD4, CDK5RAP3, MEGF10, DNAH2, PGC, DCST1, CHIA,
COL25A1, EPHX4, PKHD1, SLC2A7, ABHD1//PREB, GPNMB, CYTSA, T,
TCP11, ENAM, KDR, GPR17//LOC100291428//LIMS2, Clorf175//TTC4, IL17F,
C2orf62, GPR113//SELI, SCN10A, TRPM8, DDC//LOC100129427 and UGT3A1
(hereafter referred to interchangeably herein as "group J IRS immune system
biomarker genes" or "group J IRS biomarker genes").
[0043] In specific embodiments, the first IRS immune system biomarker is a
PLA2G7
expression product, the second IRS immune system biomarker is a PLAC8
expression
product, the third IRS immune system biomarker is a CEACAM4 expression product
and the
fourth IRS immune system biomarker is a LAMP] expression product.
[0044] In some embodiments, the indicator is for determining a likelihood of
the subject
having mild sepsis or severe sepsis, and wherein biomarker values are
determined from at
least one IRS immune system biomarker in each of first and second IRS immune
system
biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: N4BP2L2//CG030, FAM96A, MINPP1, MORC3//DOPEY2,
LSM8, PLEKHA3, MITD1, ATF4, B2M, TMX3, ZNF273, PLEKHF2, UNQ2999,
DPM1, OCLM, NADK, GPR65, SFRS3, ZNF28, PFDN4, COQ10B,
SLC30A6//DDX50, KPNA5, ATP6V1G1, HAUS2, hCG 2039148, RAB33B, BET],
UBE2V2, ATP6AP2, SUB1//TMEA1183A, TMEM188, ABHD3, LAPTM4A,
RNF138, CCDC82, TMEA1179B, PAPD4, VAMP2, CCDC126, ATG3, CHCHD5,
RBM39//L0C643167, NAT8B, HAT], CNOT6L, ZBED5, GOLT1B, TTC33,
ACTN1, ACTR6, GNB4, TMEM208, CCNH, C4orf34, HAUS6//SCARNA8,
TCTN1, SF3B14, TMEM138, TTC35, DLEU2//DLEU2L, MFSD8, COX7A2L,
UGGT2, CEPT1//DRAM2, LEAID3, CREBZF, RPL21P44, ANGEL2, UFM1,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 20 -
XPO 1, CALM2//C2orf61, PLDN, CLK1//PPIL3, Clorf84, PPA2, FPGT//TNNI3K,
ORC4L, SLC25A14, H3F3B//H3F3C, FAM188A, MCM9, KLHL9, NACA,
PAFAH1B2, CRLS1, TSSK4, LOC152217, ZNF568, ATP6V0D1//LOC 100132855,
CDC37L1, FNTA, SHFM1, JKAMP, TMFM126B, MRPL47, DENND1B,
ATP6V0E1//SNORA74B, LIN7C, HAUS3//POLN, SLC30A7, VAMP3, OBFC2A,
MAGT1, STARD3NL, C5orf15, PSMD10, RERE, RNF139, SFT2D2, SKP 1,
RNPC3//AMY2B, MYOM1, TIPRL, HPRT1, TRIM21, VRK2, CDKN1B, ANKRA2,
RAP2B, FAM127B//FAM127C//FAM127A, FAM126A, TMFM161B, UNQ1887,
FANCF, SELT, CYP20A 1, RWDD 1, ARPP 19,
SC5DL,
TICAM2//TMED7//TMED7-TICAM2, STAM, LEPROTL1, RNF44, DCP1B,
TNNT3, UCHL5, UPRT, SON, PIK3C3, SFRS1//F1J44342,
FBX022//FBX0220S, SCFD 1, Cllorf31, TMTC3,
CCDC132,
TMBIM4//L0C 100133322, ATAD1, APH1A, MYNN, HADHB, PIGN, RNA243,
5LC38A9, ClOorf84, CDKN1A, ATP7A, RPAP2, ZNF451,
GSK3B//LOC 100129275, TMSB 10, KCTD5//PR0 0461//PDPK1 , VP529,
WIPF3//ZNRF2//L0C441208, SFRS5 and Cl lorf73 (hereafter referred to
interchangeably herein as "group K IRS immune system biomarker genes" or
"group K IRS biomarker genes"); and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: Cl 3orfl, PRKCB, APOBEC3A//APOBEC3B, SFRS9,
NCAPD2//SCARNA10//FADS1, GA TS, L0C284757, TSHZ2, JAK1, MAPK13,
RPN1, GNAS, CYTSA, TRPM6, C22orf30, PCMTD1//PXDNL, CCDC69, ARSD,
MLL3//BAGE2, NCORk/C20orf191//LOC100131704, MRPS7, VEZF1 , GSR,
POU2F1, VPS4A, SMG7, PTP4A2, OSBP, GLCCI1//tcag7.903, DOCK2, PCNX,
GLTP, FBX018, YY2, TCF20, NR2C2//MRPS25, TEX2, BAMBI, WHSC1L1,
UBTF, FAH, GEMIN4, DDEF1IT1, FAM50A, VPS13D, SATB1//TBC1D5,
PARP6, SETD5, PHF21A, IRF4, ZNF217, UBE4A, HIVEP1, HDLBP, GNAI2,
MED13L, FOXP 1, NSD1, DDOST, TMBIM6, ABLIM1, SYNRG//LOC 100131822,
KDM3B, ASH1L, NCOA2, GPRIN3, NCOA1, KLF3, LOC100288114//MGC9913,
VPS26B, AHCYL1, CDC6, PLCG2, IL16, GIT2, TACC3, MAP4K4, NEK9,
FAM149B1//FAM149B2, VPS8, ATXN7, WDFY4, ZC3H11A//RP 11-74E24.2,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-21 -
THRAP3, ZNF346, AP3M2, CD14, CLASP], ABCC2,
ATXN7L1//RINT1//EFCAB10, IN080D,
CTPS,
LRRC37A3//LRRC37A2//LRRC37A//ARL 17P 1//LRRC37A4//LOC 1 00294335//L0
C644397, DNA SE], LRRN3, ZNF318, PRKAR2A, MRPS15, ANKHD1-
EIF4EBP3//ANKHD1//EIF4EBP3, BTF3L4, DGKA, ClOorf119, MBD5,
Cllorf30, CDC2L5, DPP4, DCTN4, TP53BP2, IMPDH2, GOT2, ELMO] and
PARP1 (hereafter referred to interchangeably herein as "group L IRS immune
system biomarker genes" or "group L IRS biomarker genes").
[0045] In some embodiments, the indicator is for determining a likelihood of
the subject
having mild sepsis or septic shock, and wherein biomarker values are
determined from at
least one IRS immune system biomarker in each of first and second IRS immune
system
biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: EEF1DP3, GIMAP7, ZNF839, PYGL, TNFAIP8L2, SFRS9,
VIM, GLTP, WDFY4, APPL2, C4orf3, PLD1, LIN7A, ELP2, ZDHHC3,
UBAP1//KIF24, C20orf177, FAM149B1//FAM149B2, E2F3, SPA TA], DACH1,
FAM47E//STBD1, SVIL//hCG 1783494, METTL9, LRRC42, NUPL1, UPP1,
AFF2, SLC16A4, SET, CA4, HCK, Cl6orf72, EXT1, N0P58, FRZB,
C9orf6//IKBKAP, VASP, ASB8//PHB, GTDC1, 5LC39A9, FBX034//KIAA0831,
RABGEF1//tcag7.967//tcag7.951//KCTD7//LOC100293333,
5LC28A3,
WIPI1//ARSG, NFE2, GOLGA1//SCAI, C9orf84, RPS6KA2, PSMA7, Cl9orf59,
ICA1, TOR1AIP2//TOR1AIP1//IFRG15, MSRA, FPR1, TP53I3, FOXL2, CD63,
PIGC, CENPBD1, CYB5R1, GNB2, ZNF197, KLF7, NSFL1C, USF2, PARP6,
MAP9, TSPO, CSTB, DDA1, 5LC36A4, GFOD2, OCRL, ZNF232, APH1B,
TALD01//INTS8, DENND2C//BCAS2, RAB11FIP2, LAPS, PLP2, EIF4E2,
DNASE1L1//RPL10, AFTPH, TMC03, RPA2, UQCRC1, ZDHHC3, and ACTR1A
(hereafter referred to interchangeably herein as "group M IRS immune system
biomarker genes" or "group M IRS biomarker genes"); and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: CD6, ITPA, PVRIG//PILRB//STAG3, FLT3LG, IL12RB1,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 22 -
M4P3K14//L0C100133991, FAM102A, TMC8, TMEA1208, TMEM109, C1orf84,
NADK, SEPT], UBA7, CD5, Cl2orf62, C20orf112, FOXP4, EIF4A2//SNORA4,
ZNF4871/L0C439911, KCTD13, IL18BP//NUMA1, KPNA5, EDC4,
ZNF587//ZNF417, NBR2, RPL28, ZNF738, SHFM1, CNO, C9orf82,
RPL5//SNORA66//SNORD21//FAM69A, VAMP2, SIT], SFRS3, LCK, IRF9,
MRPS21, NEFM//LOC100129717, RCN2, BET], Cl9orf6, SH2D1A, GLS,
OR1C1, RBM14//RBM4, CCDC97, TUT], MRPL14, ENOPH1, NAGK, DPM1,
MPV17, SH2D3C, TMEA1204, C3orf42, ARSD, FAM96A, LSM8, ATP5G1,
KTI12, ARL4C, Cllorf31,
Cl6orf58//LOC100128371,
CCDC109B//HIGD1A//CCDC13, NUDT21, ZFP106, ACTR6, LIX1L, MEF2D,
ZNF407, TMEM18, NAT]], DNAJC24, PLEKHA3, GPN2, SMCR7,
C7orf23//DMTF1, PPM1G, CNOT6L, NA CA, FNTA, GRIA2, N4BP2L2//CG030,
ENOSF1//TYMS, THBS4, LUC7L2, MOCS2, ZNF383, AKNA, UBE2Z, FLI34077,
SH3KBP1, POLR2F//LOC100131530, ILIA, UBE2V2, KIAA1919, PRKCB,
SHOC2, RBM46, GRPEL2, KCNG3, PCDH10, XAB2, VP552, MCCC2,
NSMCE4A, PTP4A2, SNX2, COQ10A, C6orf182, RNF44, MOGS, DIRAS3,
Mitochondrial, KIAA1826, SGK196, NSUN5//NSUN5B//NSUN5C,Mitochondrial,
MORF4L2, MAK16//C8orf41, PILRB//PVRIG//STAG3, SAAL1, TMX3, PTPRG,
MAPK1, DNAJA3, LEAP2, LMOD3, ASB6, MTMR10//MTMR15, HIBADH,
MORC1, CORO1A//L0C606724, SFXN2, HSN2, AAAS, INHBA, MRPS7,
LRRFIP1, KCTD7//RABGEF1, DCDC2//KAAG1, SLCO3A1, DENND4B, CFTR,
MOG, QRFPR, BAT2//SNORA38, ITPR3, C3orf22, TNFRSF4, ZNF646//ZNF668,
GCM2, CDK4//TSPAN31, FXCl, RNMT, CHST4, POLR3E, PDE8B, C6orf146,
CHCHD5, TARBP1, TADA1L, PREX2, TRAP1//DNASE1, ZNHIT6,
RGP1//GBA2, SMCP, ZC3H15, TRAPPC4, SARNP//DNAJC14, GNAS,
Cl4orf104, IL2ORA, WAC, SLIT3, C4orf39, TSEN54, PPP1R13B, TRIM35,
HGC6.3, YIPF3, HQ0644/PR00644, FAM13C//PHYHIPL, BCCIP//DHX32,
ACADM, SUB1//TMEM183A, KPNA 1, SPAG17//WDR3, KI4A1549,
DIABLO//B3GNT4, ZBTB44, EIF1AD, RUNX1T1, TRIL, PTPLB, DHDDS,
UPK1A, OTUB1, Clorf182, HAPLN2, SOBP, RYR3, LRRC17, TKTL2, TMBIM6,
and GDNF (hereafter referred to interchangeably herein as "group N IRS immune
system biomarker genes" or "group N IRS biomarker genes").

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 23 -
[0046] In some embodiments, the indicator is for determining a likelihood of
the subject
having severe sepsis or septic shock, and wherein biomarker values are
determined from at
least one IRS immune system biomarker in each of first and second IRS immune
system
biomarker groups, wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: SIRPG // SIRPA, GATA3, FAM102A, UPF3A, ATP13A5,
CACNA1I and RANBP17 // USP12 (hereafter referred to interchangeably herein as
"group 0 IRS immune system biomarker genes" or "group 0 IRS biomarker
genes"); and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from the following IRS immune system
biomarker genes: GABRA6, HAPLN1, YSK4, FOXL2, TLL1, MECOM, COL3A1,
HRG, SLC22A3, C8orf45, SCN7A and SNTG1 (hereafter referred to
interchangeably herein as "group P IRS immune system biomarker genes" or
"group P IRS biomarker genes").
[0047] In another broad form the present invention seeks to provide apparatus
for
determining an indicator used in assessing a likelihood of a biological
subject having a
presence, absence, degree or prognosis of at least one medical condition, the
apparatus
including at least one electronic processing device that:
a) determines a pair of biomarker values, each biomarker value being a value
measured or derived for at least one corresponding immune system biomarker of
the biological subject and being at least partially indicative of a
concentration of
the immune system biomarker in a sample taken from the subject;
b) determines a derived biomarker value using the pair of biomarker values,
the
derived biomarker value being indicative of a ratio of concentrations of the
pair of
immune system biomarkers; and,
c) determines the indicator using the derived biomarker value.
[0048] In another broad form the present invention seeks to provide a
composition
comprising at least one pair of reverse transcribed mRNAs and at least one
oligonucleotide
primer or probe that hybridizes to an individual one of the reverse
transcribed mRNAs, the at

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 24 -
least one pair of reverse transcribed mRNAs comprising a first pair and a
second pair of
reverse transcribed mRNAs, wherein the first pair comprises a PLAC8 reverse
transcribed
mRNA and a PLA2G7 reverse transcribed mRNA and wherein the second pair
comprises a
CEACAM4 reverse transcribed mRNA and a LAMP] reverse transcribed mRNA.
[0049] In another broad form the present invention seeks to provide a
composition
comprising at least one pair of reverse transcribed mRNAs and at least one
oligonucleotide
primer or probe that hybridizes to an individual one of the reverse
transcribed mRNAs, the at
least one pair of reverse transcribed mRNAs comprising a reverse transcribed
mRNA from a
first IRS immune system biomarker gene selected from group A IRS immune system

biomarker genes and a reverse transcribed mRNA from a second IRS immune system

biomarker gene selected from group B IRS immune system biomarker genes.
[0050] In another broad form the present invention seeks to provide a
composition
comprising at least one pair of reverse transcribed mRNAs and at least one
oligonucleotide
primer or probe that hybridizes to an individual one of the reverse
transcribed mRNAs, the at
least one pair of reverse transcribed mRNAs comprising a reverse transcribed
mRNA from a
first IRS immune system biomarker gene selected from group C IRS immune system

biomarker genes and a reverse transcribed mRNA from a second IRS immune system

biomarker gene selected from group D IRS immune system biomarker genes.
[0051] In another broad form the present invention seeks to provide a
composition
comprising at least one pair of reverse transcribed mRNAs and at least one
oligonucleotide
primer or probe that hybridizes to an individual one of the reverse
transcribed mRNAs, the at
least one pair of reverse transcribed mRNAs comprising a reverse transcribed
mRNA from a
first IRS immune system biomarker gene selected from group E IRS immune system

biomarker genes and a reverse transcribed mRNA from a second IRS immune system

biomarker gene selected from group F IRS immune system biomarker genes.
[0052] In another broad form the present invention seeks to provide a
composition
comprising at least two pairs of reverse transcribed mRNAs and at least one
oligonucleotide
primer or probe that hybridizes to an individual one of the reverse
transcribed mRNAs, the at
least two pairs of reverse transcribed mRNAs comprising a first pair and a
second pair of
reverse transcribed mRNAs, wherein the first pair comprises a reverse
transcribed mRNA
from a first IRS immune system biomarker gene and a reverse transcribed mRNA
from a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 25 -
second IRS immune system biomarker gene, and wherein the second pair comprises
a reverse
transcribed mRNA from a third IRS immune system biomarker gene and a reverse
transcribed mRNA from a fourth IRS immune system biomarker gene, wherein the
first IRS
immune system biomarker gene is selected from group G IRS immune system
biomarker
genes, wherein the second IRS immune system biomarker gene is selected from
group H IRS
immune system biomarker genes, wherein the third IRS immune system biomarker
gene is
selected from group I IRS immune system biomarker genes, and wherein the
fourth IRS
immune system biomarker gene is selected from group J IRS immune system
biomarker
genes.
[0053] In another broad form the present invention seeks to provide a
composition
comprising at least one pair of reverse transcribed mRNAs and at least one
oligonucleotide
primer or probe that hybridizes to an individual one of the reverse
transcribed mRNAs, the at
least one pair of reverse transcribed mRNAs comprising a reverse transcribed
mRNA from a
first IRS immune system biomarker gene selected from group K IRS immune system

biomarker genes and a reverse transcribed mRNA from a second IRS immune system

biomarker gene selected from group L IRS immune system biomarker genes.
[0054] In another broad form the present invention seeks to provide a
composition
comprising at least one pair of reverse transcribed mRNAs and at least one
oligonucleotide
primer or probe that hybridizes to an individual one of the reverse
transcribed mRNAs, the at
least one pair of reverse transcribed mRNAs comprising a reverse transcribed
mRNA from a
first IRS immune system biomarker gene selected from group M IRS immune system

biomarker genes and a reverse transcribed mRNA from a second IRS immune system

biomarker gene selected from group N IRS immune system biomarker genes.
[0055] In another broad form the present invention seeks to provide a
composition
comprising at least one pair of reverse transcribed mRNAs and at least one
oligonucleotide
primer or probe that hybridizes to an individual one of the reverse
transcribed mRNAs, the at
least one pair of reverse transcribed mRNAs comprising a reverse transcribed
mRNA from a
first IRS immune system biomarker gene selected from group 0 IRS immune system

biomarker genes and a reverse transcribed mRNA from a second IRS immune system

biomarker gene selected from group P IRS immune system biomarker genes.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 26 -
[0056] The at least one oligonucleotide primer or probe can be hybridized to
an individual
one of the reverse transcribed mRNAs.
[0057] The reverse transcribed mRNAs can be derived from components of the
immune
system.
[0058] The reverse transcribed mRNAs can be derived from leukocytes.
[0059] The reverse transcribed mRNAs can be derived from blood cells.
[0060] The reverse transcribed mRNAs can be derived from peripheral blood
cells.
[0061] The composition can further comprise a labeled reagent for detecting
the reverse
transcribed mRNAs.
[0062] The labeled reagent can be a labeled said at least one oligonucleotide
primer or probe.
[0063] The labeled reagent can be a labeled said reverse transcribed mRNA.
[0064] The labeled reagent can be a labeled oligonucleotide linker or tag for
labeling a said
reverse transcribed mRNA.
[0065] In another broad form the present invention seeks to provide a kit for
determining an
indicator indicative of the likelihood of the presence or absence of at least
one condition
selected from the group consisting of inSIRS and ipSIRS, the kit comprising at
least one pair
of reagents comprising a first pair of reagents and a second pair of reagents,
wherein the first
pair of reagents comprises (i) a reagent that allows quantification of a
polynucleotide
expression product of the PLA2G7 gene; and (ii) a reagent that allows
quantification of a
polynucleotide expression product of the PLAC8 gene, wherein the second pair
of reagents
comprises: (iii) a reagent that allows quantification of a polynucleotide
expression product of
the CEACAM4 gene; and (iv) a reagent that allows quantification of a
polynucleotide
expression product of the LAMP] gene.
[0066] In another broad form the present invention seeks to provide a kit for
determining an
indicator indicative of the likelihood of the presence or absence of at least
one condition
selected from the group consisting of inSIRS and a healthy condition, the kit
comprising at
least one pair of reagents comprising (i) a reagent that allows quantification
of a
polynucleotide expression product of a first IRS immune system biomarker gene;
and (ii) a
reagent that allows quantification of a polynucleotide expression product of a
second IRS

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 27 -
immune system biomarker gene, wherein the first IRS immune system biomarker
gene is
selected from group A IRS immune system biomarker genes and wherein the second
IRS
immune system biomarker gene is selected from group B IRS immune system
biomarker
genes.
[0067] In another broad form the present invention seeks to provide a kit for
determining an
indicator indicative of the likelihood of the presence or absence of at least
one condition
selected from the group consisting of ipSIRS and a healthy condition, the kit
comprising at
least one pair of reagents comprising (i) a reagent that allows quantification
of a
polynucleotide expression product of a first IRS immune system biomarker gene;
and (ii) a
reagent that allows quantification of a polynucleotide expression product of a
second IRS
immune system biomarker gene, wherein the first IRS immune system biomarker
gene is
selected from group C IRS immune system biomarker genes and wherein the second
IRS
immune system biomarker gene is selected from group D IRS immune system
biomarker
genes.
[0068] In another broad form the present invention seeks to provide a kit for
determining an
indicator indicative of the likelihood of the presence or absence of at least
one condition
selected from the group consisting of inSIRS and ipSIRS, the kit comprising at
least one pair
of reagents comprising (i) a reagent that allows quantification of a
polynucleotide expression
product of a first IRS immune system biomarker gene; and (ii) a reagent that
allows
quantification of a polynucleotide expression product of a second IRS immune
system
biomarker gene, wherein the first IRS immune system biomarker gene is selected
from group
E IRS immune system biomarker genes and wherein the second IRS immune system
biomarker gene is selected from group F IRS immune system biomarker genes.
[0069] In another broad form the present invention seeks to provide a kit for
determining an
indicator indicative of the likelihood of the presence or absence of at least
one condition
selected from the group consisting of inSIRS and ipSIRS, the kit comprising at
least two
pairs of reagents comprising a first pair of reagents and a second pair of
reagents, wherein the
first pair of reagents comprises (i) a reagent that allows quantification of a
polynucleotide
expression product of a first IRS immune system biomarker gene; and (ii) a
reagent that
allows quantification of a polynucleotide expression product of a second IRS
immune system
biomarker gene, and wherein the second pair of reagents comprises (i) a
reagent that allows

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 28 -
quantification of a polynucleotide expression product of a third IRS immune
system
biomarker gene; and (ii) a reagent that allows quantification of a
polynucleotide expression
product of a fourth IRS immune system biomarker gene, wherein the first IRS
immune
system biomarker gene is selected from group G IRS immune system biomarker
genes,
wherein the second IRS immune system biomarker gene is selected from group H
IRS
immune system biomarker genes, wherein the third IRS immune system biomarker
gene is
selected from group I IRS immune system biomarker genes, and wherein the
fourth IRS
immune system biomarker gene is selected from group J IRS immune system
biomarker
genes.
[0070] In another broad form the present invention seeks to provide a kit for
determining an
indicator indicative of the likelihood of the presence or absence of at least
one condition
selected from the group consisting of mild sepsis and severe sepsis, the kit
comprising at least
one pair of reagents comprising (i) a reagent that allows quantification of a
polynucleotide
expression product of a first IRS immune system biomarker gene; and (ii) a
reagent that
allows quantification of a polynucleotide expression product of a second IRS
immune system
biomarker gene, wherein the first IRS immune system biomarker gene is selected
from group
K IRS immune system biomarker genes and wherein the second IRS immune system
biomarker gene is selected from group L IRS immune system biomarker genes.
[0071] In another broad form the present invention seeks to provide a kit for
determining an
indicator indicative of the likelihood of the presence or absence of at least
one condition
selected from the group consisting of mild sepsis and septic shock, the kit
comprising at least
one pair of reagents comprising (i) a reagent that allows quantification of a
polynucleotide
expression product of a first IRS immune system biomarker gene; and (ii) a
reagent that
allows quantification of a polynucleotide expression product of a second IRS
immune system
biomarker gene, wherein the first IRS immune system biomarker gene is selected
from group
M IRS immune system biomarker genes and wherein the second IRS immune system
biomarker gene is selected from group N IRS immune system biomarker genes.
[0072] In another broad form the present invention seeks to provide a kit for
determining an
indicator indicative of the likelihood of the presence or absence of at least
one condition
selected from the group consisting of severe sepsis and septic shock, the kit
comprising at
least one pair of reagents comprising (i) a reagent that allows quantification
of a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 29 -
polynucleotide expression product of a first IRS immune system biomarker gene;
and (ii) a
reagent that allows quantification of a polynucleotide expression product of a
second IRS
immune system biomarker gene, wherein the first IRS immune system biomarker
gene is
selected from group 0 IRS immune system biomarker genes and wherein the second
IRS
immune system biomarker gene is selected from group P IRS immune system
biomarker
genes.
[0073] In another broad form the present invention seeks to provide a method
for inhibiting
the development or progression in a subject of at least one condition selected
from the group
consisting of inSIRS and ipSIRS, the method comprising: exposing the subject
to a treatment
regimen for treating the at least one condition based on an indicator obtained
from an
indicator-determining method, wherein the indicator is indicative of the
presence of the at
least one condition in the subject, the indicator-determining method
comprising: (a)
determining at least one pair of biomarker values, each biomarker value being
a value
measured or derived for at least one corresponding immune system biomarker of
the
biological subject and being at least partially indicative of a concentration
of the immune
system biomarker in a sample taken from the subject, (b) determining at least
one derived
biomarker value using the at least one pair of biomarker values, the derived
biomarker value
being indicative of a ratio of concentrations of the at least one pair of
immune system
biomarkers; and (c) determining the indicator based on the at least one
derived biomarker
value, wherein the pair of biomarker values comprises at least one of:
a) a first pair of biomarker values comprising first and second biomarker
values
corresponding to first and second biomarkers, wherein the first immune system
biomarker represents a polynucleotide expression product of the PLA2G7 gene
and wherein the second immune system biomarker representing a polynucleotide
expression product of the PLAC8 gene, and
b) a second pair of biomarker values comprises third and fourth biomarker
values
corresponding to third and fourth immune system biomarkers, respectively,
wherein the third immune system biomarker represents a polynucleotide
expression product of the CEACAM4 gene and wherein the fourth immune system
biomarker represents a polynucleotide expression product of the LAMP] gene.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 30 -
[0074] Typically the indicator-determining method comprises: determining the
first pair and
second pair of biomarker values and determining a first derived biomarker
value calculated
using the first pair of biomarker values and a second derived biomarker value
calculated
using the second pair of biomarker values; and determining the indicator based
on a
combination of the first and second derived biomarker values.
[0075] In another broad form the present invention seeks to provide a method
for inhibiting
the development or progression of inSIRS in a subject, the method comprising:
exposing the
subject to a treatment regimen for treating inSIRS based on an indicator
obtained from an
indicator-determining method, wherein the indicator is indicative of the
presence of inSIRS
in the subject, the indicator-determining method comprising: (a) determining
at least one pair
of biomarker values, each biomarker value being a value measured or derived
for at least one
corresponding immune system biomarker of the biological subject and being at
least partially
indicative of a concentration of the immune system biomarker in a sample taken
from the
subject, (b) determining at least one derived biomarker value using the at
least one pair of
biomarker values, the derived biomarker value being indicative of a ratio of
concentrations of
the pair of immune system biomarkers; and (c) determining the indicator based
on the at least
one derived biomarker value, wherein the at least one pair of biomarker values
comprises
first and second biomarker values corresponding to first and second immune
system
biomarkers, respectively, wherein the first immune system biomarker represents
a
polynucleotide expression product of a first IRS immune system biomarker gene,
and
wherein the second immune system biomarker represents a polynucleotide
expression
product of a second IRS immune system biomarker gene, wherein the first IRS
immune
system biomarker gene is selected from group A IRS immune system biomarker
genes and
wherein the second IRS immune system biomarker gene is selected from group B
IRS
immune system biomarker genes.
[0076] In another broad form the present invention seeks to provide a method
for inhibiting
the development or progression of ipSIRS in a subject, the method comprising:
exposing the
subject to a treatment regimen for treating ipSIRS based on an indicator
obtained from an
indicator-determining method, wherein the indicator is indicative of the
presence of ipSIRS
in the subject, the indicator-determining method comprising: (a) determining
at least one pair
of biomarker values, each biomarker value being a value measured or derived
for at least one

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 3 1 -
corresponding immune system biomarker of the biological subject and being at
least partially
indicative of a concentration of the immune system biomarker in a sample taken
from the
subject, (b) determining at least one derived biomarker value using the at
least one pair of
biomarker values, the derived biomarker value being indicative of a ratio of
concentrations of
the at least one pair of immune system biomarkers; and (c) determining the
indicator based
on the at least one derived biomarker value, wherein the at least one pair of
biomarker values
comprises first and second biomarker values corresponding to first and second
immune
system biomarkers, respectively, wherein the first immune system biomarker
represents a
polynucleotide expression product of a first IRS immune system biomarker gene,
and
wherein the second immune system biomarker represents a polynucleotide
expression
product of a second IRS immune system biomarker gene, wherein the first IRS
immune
system biomarker gene is selected from group C IRS immune system biomarker
genes and
wherein the second IRS immune system biomarker gene is selected from group D
IRS
immune system biomarker genes.
[0077] In another broad form the present invention seeks to provide a method
for inhibiting
the development or progression in a subject of at least one condition selected
from the group
consisting of inSIRS and ipSIRS, the method comprising: exposing the subject
to a treatment
regimen for treating the at least one condition based on an indicator obtained
from an
indicator-determining method, wherein the indicator is indicative of the
presence of the at
least one condition in the subject, the indicator-determining method
comprising: (a)
determining at least one pair of biomarker values, each biomarker value being
a value
measured or derived for at least one corresponding immune system biomarker of
the
biological subject and being at least partially indicative of a concentration
of the immune
system biomarker in a sample taken from the subject, (b) determining at least
one derived
biomarker value using the at least one pair of biomarker values, the derived
biomarker value
being indicative of a ratio of concentrations of the at least one pair of
immune system
biomarkers; and (c) determining the indicator based on the at least one
derived biomarker
value, wherein the at least one pair of biomarker values comprises first and
second biomarker
values corresponding to of first and second immune system biomarkers,
respectively,
wherein the first immune system biomarker represents a polynucleotide
expression product
of a first IRS immune system biomarker gene, and wherein the second immune
system
biomarker represents a polynucleotide expression product of a second IRS
immune system

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 32 -
biomarker gene, wherein the first IRS immune system biomarker gene is selected
from group
E IRS immune system biomarker genes and wherein the second IRS immune system
biomarker gene is selected from group F IRS immune system biomarker genes.
[0078] In another broad form the present invention seeks to provide a method
for inhibiting
the development or progression in a subject of at least one condition selected
from the group
consisting of inSIRS and ipSIRS, the method comprising: exposing the subject
to a treatment
regimen for treating the at least one condition based on an indicator obtained
from an
indicator-determining method, wherein the indicator is indicative of the
presence of the at
least one condition in the subject, the indicator-determining method
comprising: (a)
determining at least two pairs of biomarker values, each biomarker value being
a value
measured or derived for at least one corresponding immune system biomarker of
the
biological subject and being at least partially indicative of a concentration
of the immune
system biomarker in a sample taken from the subject, (b) determining at least
two derived
biomarker values using the at least two pairs of biomarker values, the derived
biomarker
value being indicative of a ratio of concentrations of each pair of immune
system biomarkers;
and (c) determining the indicator based on the at least two derived biomarker
values, wherein
the at least one pair of biomarker values comprises a first pair of biomarker
values
comprising first and second biomarker values corresponding to first and second
immune
system biomarkers, respectively, wherein the first immune system biomarker
represents a
polynucleotide expression product of a first IRS immune system biomarker gene
and wherein
the second immune system biomarker represents a polynucleotide expression
product of a
second IRS immune system biomarker gene, and a second pair of biomarker values

comprising third and fourth biomarker values corresponding to third and fourth
immune
system biomarkers, respectively, wherein the third immune system biomarker
represents a
polynucleotide expression product of a third IRS immune system biomarker gene
and
wherein the fourth immune system biomarker represents a polynucleotide
expression product
of a fourth IRS immune system biomarker gene, wherein the first IRS immune
system
biomarker gene is selected from group G IRS immune system biomarker genes,
wherein the
second IRS immune system biomarker gene is selected from group H IRS immune
system
biomarker genes, wherein the third IRS immune system biomarker gene is
selected from
group I IRS immune system biomarker genes, and wherein the fourth IRS immune
system
biomarker gene is selected from group J IRS immune system biomarker genes.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 33 -
[0079] In another broad form the present invention seeks to provide a method
for inhibiting
the development or progression in a subject of at least one condition selected
from the group
consisting of mild sepsis and severe sepsis, the method comprising: exposing
the subject to a
treatment regimen for treating the at least one condition based on an
indicator obtained from
an indicator-determining method, wherein the indicator is indicative of the
presence of the at
least one condition in the subject, the indicator-determining method
comprising: (a)
determining at least one pair of biomarker values, each biomarker value being
a value
measured or derived for at least one corresponding immune system biomarker of
the
biological subject and being at least partially indicative of a concentration
of the immune
system biomarker in a sample taken from the subject, (b) determining at least
one derived
biomarker value using the at least one pair of biomarker values, the derived
biomarker value
being indicative of a ratio of concentrations of the at least one pair of
immune system
biomarkers; and (c) determining the indicator based on the at least one
derived biomarker
value, wherein the at least one pair of biomarker values comprises first and
second biomarker
values corresponding to first and second immune system biomarkers,
respectively, wherein
the first immune system biomarker represents a polynucleotide expression
product of a first
IRS immune system biomarker gene, and wherein the second immune system
biomarker
represents a polynucleotide expression product of a second IRS immune system
biomarker
gene, wherein the first IRS immune system biomarker gene is selected from
group K IRS
immune system biomarker genes and wherein the second IRS immune system
biomarker
gene is selected from group L IRS immune system biomarker genes.
[0080] In another broad form the present invention seeks to provide a method
for inhibiting
the development or progression in a subject of at least one condition selected
from the group
consisting of mild sepsis and septic shock, the method comprising: exposing
the subject to a
treatment regimen for treating the at least one condition based on an
indicator obtained from
an indicator-determining method, wherein the indicator is indicative of the
presence of the at
least one condition in the subject, the indicator-determining method
comprising: (a)
determining at least one pair of biomarker values, each biomarker value being
a value
measured or derived for at least one corresponding immune system biomarker of
the
biological subject and being at least partially indicative of a concentration
of the immune
system biomarker in a sample taken from the subject, (b) determining at least
one derived
biomarker value using the at least one pair of biomarker values, the derived
biomarker value

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 34 -
being indicative of a ratio of concentrations of the at least one pair of
immune system
biomarkers; and (c) determining the indicator based on the at least one
derived biomarker
value, wherein the at least one pair of biomarker values comprises first and
second biomarker
values corresponding to first and second immune system biomarkers,
respectively, wherein
the first immune system biomarker represents a polynucleotide expression
product of a first
IRS immune system biomarker gene, and wherein the second immune system
biomarker
represents a polynucleotide expression product of a second IRS immune system
biomarker
gene, wherein the first IRS immune system biomarker gene is selected from
group M IRS
immune system biomarker genes and wherein the second IRS immune system
biomarker
gene is selected from group N IRS immune system biomarker genes.
[0081] In another broad form the present invention seeks to provide a method
for inhibiting
the development or progression in a subject of at least one condition selected
from the group
consisting of severe sepsis and septic shock, the method comprising: exposing
the subject to
a treatment regimen for treating the at least one condition based on an
indicator obtained
from an indicator-determining method, wherein the indicator is indicative of
the presence of
the at least one condition in the subject, the indicator-determining method
comprising: (a)
determining at least one pair of biomarker values, each biomarker value being
a value
measured or derived for at least one corresponding immune system biomarker of
the
biological subject and being at least partially indicative of a concentration
of the immune
system biomarker in a sample taken from the subject, (b) determining at least
one derived
biomarker value using the at least one pair of biomarker values, the derived
biomarker value
being indicative of a ratio of concentrations of the at least one pair of
immune system
biomarkers; and (c) determining the indicator based on the at least one
derived biomarker
value, wherein the at least one pair of biomarker values comprises first and
second biomarker
values corresponding to first and second immune system biomarkers,
respectively, wherein
the first immune system biomarker represents a polynucleotide expression
product of a first
IRS immune system biomarker gene, and wherein the second immune system
biomarker
represents a polynucleotide expression product of a second IRS immune system
biomarker
gene, wherein the first IRS immune system biomarker gene is selected from
group 0 IRS
immune system biomarker genes and wherein the second IRS immune system
biomarker
gene is selected from group P IRS immune system biomarker genes.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 35 -
[0082] In some embodiments, the method comprises taking the sample from the
subject and
obtaining the indicator according to the indicator-determining method.
[0083] In some embodiments, the method comprises: sending the sample taken
from the
subject to a laboratory at which the indicator is determined.
[0084] Typically, the sample comprises cells obtained from the subject or a
nucleic acid
sample thereof.
[0085] In another broad form the present invention seeks to provide a method
for
differentiating between inSIRS and ipSIRS in a biological subject, the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a
pair of biomarker values, the pair of biomarker values being selected from the

group consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the PLA2G7 gene and PLAC8 gene;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide expression products of the CEACAM4 gene and LAMP] gene;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of inSIRS and ipSIRS,
respectively;
and,
e) determining a likelihood of the subject having inSIRS or ipSIRS in
accordance
with the results of the comparison.
[0086] In another broad form the present invention seeks to provide a method
for
differentiating between inSIRS and a healthy condition in a biological
subject, the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a
pair of biomarker values, the pair of biomarker values being indicative of a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 36 -
concentration of polynucleotide expression products of a first IRS immune
system
biomarker gene and a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group A IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group B IRS immune system biomarker genes;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of inSIRS and healthy condition,
respectively; and,
e) determining a likelihood of the subject having inSIRS or the healthy
condition in
accordance with the results of the comparison.
[0087] In another broad form the present invention seeks to provide a method
for
differentiating between ipSIRS and a healthy condition in a biological
subject, the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a
pair of biomarker values, the pair of biomarker values being indicative of a
concentration of polynucleotide expression products of a first IRS immune
system
biomarker gene and a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group C IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group D IRS immune system biomarker genes;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of ipSIRS and healthy condition,
respectively; and,
e) determining a likelihood of the subject having ipSIRS or the healthy
condition in
accordance with the results of the comparison.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 37 -
[0088] In another broad form the present invention seeks to provide a method
for
differentiating between inSIRS and ipSIRS in a biological subject, the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a
pair of biomarker values, the pair of biomarker values being indicative of a
concentration of polynucleotide expression products of a first IRS immune
system
biomarker gene and a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group E IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group F IRS immune system biomarker genes;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of inSIRS and ipSIRS,
respectively;
and,
e) determining a likelihood of the subject having inSIRS or ipSIRS in
accordance
with the results of the comparison.
[0089] In another broad form the present invention seeks to provide a method
for
differentiating between inSIRS and ipSIRS in a biological subject, the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a
pair of biomarker values, the pair of biomarker values being selected from the

group consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of a first IRS immune system biomarker gene and a
second IRS immune system biomarker gene, wherein the first IRS immune
system biomarker gene is selected from group G IRS immune system
biomarker genes and wherein the second IRS immune system biomarker gene
is selected from group H IRS immune system biomarker genes;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 38 -
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide expression products of a third IRS immune system biomarker
gene and a fourth IRS immune system biomarker gene, wherein the third IRS
immune system biomarker gene is selected from group I IRS immune system
biomarker genes and wherein the fourth IRS immune system biomarker gene
is selected from group J IRS immune system biomarker genes;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of inSIRS and ipSIRS,
respectively;
and,
e) determining a likelihood of the subject having inSIRS or ipSIRS in
accordance
with the results of the comparison.
[0090] In another broad form the present invention seeks to provide a method
for
differentiating between mild sepsis and severe sepsis in a biological subject,
the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a
pair of biomarker values, the pair of biomarker values being indicative of a
concentration of polynucleotide expression products of a first IRS immune
system
biomarker gene and a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group K IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group L IRS immune system biomarker genes;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of mild sepsis and severe sepsis,

respectively; and,
e) determining a likelihood of the subject having mild sepsis or severe sepsis
in
accordance with the results of the comparison.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 39 -
[0091] In another broad form the present invention seeks to provide a method
for
differentiating between mild sepsis and septic shock in a biological subject,
the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a
pair of biomarker values, the pair of biomarker values being indicative of a
concentration of polynucleotide expression products of a first IRS immune
system
biomarker gene and a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group M IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group N IRS immune system biomarker genes;
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of mild sepsis and septic shock,
respectively; and,
e) determining a likelihood of the subject having mild sepsis or septic shock
in
accordance with the results of the comparison.
[0092] In another broad form the present invention seeks to provide a method
for
differentiating between severe sepsis and septic shock in a biological
subject, the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) quantifying polynucleotide expression products within the sample to
determine a
pair of biomarker values, the pair of biomarker values being indicative of a
concentration of polynucleotide expression products of a first IRS immune
system
biomarker gene and a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group 0 IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group P IRS immune system biomarker genes;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 40 -
c) determining an indicator indicative of a ratio of concentrations of the
polynucleotide expression products using the pair of biomarker values; and,
d) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of severe sepsis and septic
shock,
respectively; and,
e) determining a likelihood of the subject having severe sepsis or septic
shock in
accordance with the results of the comparison.
[0093] Typically the method includes determining:
a) a first derived biomarker value indicative of a ratio of concentrations of
the
polynucleotide expression products using the first pair of biomarker values;
b) a second derived biomarker value indicative of a ratio of concentrations of
the
polynucleotide expression products using the first pair of biomarker values;
and,
c) determining the indicator by combining the first and second derived
biomarker
values.
[0094] Typically the first and second indicator references are distributions
of indicators
determined for first and second groups of a reference population, the first
and second group
consisting of individuals diagnosed with inSIRS and ipSIRS respectively.
[0095] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing the likelihood of a biological
subject having at
least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products selected from the group consisting of:
i) a first pair of polynucleotide expression products of the PLA2G7 gene and
PLAC8 gene;
ii) a second pair of polynucleotide expression products of the CEACAM4 gene
and LAMP] gene;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-41 -
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a
medical condition.
[0096] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing the likelihood of a biological
subject having at
least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products selected from the group consisting of: a polynucleotide expression
product of a first IRS immune system biomarker gene and a polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group A IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group B IRS immune system biomarker genes;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a
medical condition.
[0097] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing the likelihood of a biological
subject having at
least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products selected from the group consisting of: a polynucleotide expression

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 42 -
product of a first IRS immune system biomarker gene and a polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group C IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group D IRS immune system biomarker genes;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a
medical condition.
[0098] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing the likelihood of a biological
subject having at
least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products selected from the group consisting of: a polynucleotide expression
product of a first IRS immune system biomarker gene and a polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group E IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group F IRS immune system biomarker genes;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a
medical condition.
[0099] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing the likelihood of a biological
subject having at
least one medical condition, the method including:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 43 -
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products selected from the group consisting of:
i) a first pair of polynucleotide expression products of a first IRS immune
system biomarker gene and a second IRS immune system biomarker gene,
wherein the first IRS immune system biomarker gene is selected from group G
IRS immune system biomarker genes and wherein the second IRS immune
system biomarker gene is selected from group H IRS immune system
biomarker genes;
ii) a second pair of polynucleotide expression products of a third IRS immune
system biomarker gene and a fourth IRS immune system biomarker gene,
wherein the third IRS immune system biomarker gene is selected from group I
IRS immune system biomarker genes and wherein the fourth IRS immune
system biomarker gene is selected from group J IRS immune system
biomarker genes;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a
medical condition.
[0100] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing the likelihood of a biological
subject having at
least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products selected from the group consisting of: a polynucleotide expression
product of a first IRS immune system biomarker gene and a polynucleotide

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 44 -
expression product of a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group K IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group L IRS immune system biomarker genes;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a
medical condition.
[0101] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing the likelihood of a biological
subject having at
least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products selected from the group consisting of: a polynucleotide expression
product of a first IRS immune system biomarker gene and a polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
second IRS immune system biomarker gene is selected from group M IRS
immune system biomarker genes and wherein the second IRS immune system
biomarker gene is selected from group N IRS immune system biomarker genes;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a
medical condition.
[0102] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing the likelihood of a biological
subject having at
least one medical condition, the method including:
a) obtaining a sample taken from a biological subject, the sample including
polynucleotide expression products;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 45 -
b) amplifying at least some polynucleotide expression products in the sample;
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products selected from the group consisting of: a polynucleotide expression
product of a first IRS immune system biomarker gene and a polynucleotide
expression product of a second IRS immune system biomarker gene, wherein the
first IRS immune system biomarker gene is selected from group 0 IRS immune
system biomarker genes and wherein the second IRS immune system biomarker
gene is selected from group P IRS immune system biomarker genes;
d) determining the indicator by determining a difference between the
amplification
amounts; and,
e) using the indicator to assess the likelihood of a biological subject having
a
medical condition.
[0103] Typically the method includes determining:
a) a first derived biomarker value by determining a difference between the
amplification amounts of the first pair of polynucleotide expression products;
b) a second derived biomarker value by determining a difference between the
amplification amounts of the second pair of polynucleotide expression
products;
c) determining the indicator by adding the first and second derived biomarker
values.
[0104] Typically the method includes determining:
a) comparing the indicator to first and second indicator references, wherein
the first
and second indicator references are distributions of indicators determined for
first
and second groups of a reference population, one of the first and second
groups
consisting of individuals diagnosed with the medical condition; and,
b) determining a likelihood of the subject having the medical condition in
accordance with the results of the comparison.
[0105] Typically the amplification amount is at least one of:
a) a cycle time;
b) a number of cycles;
c) a cycle threshold;
d) an amplification time; and,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 46 -
e) relative to an amplification amount of another amplified product.
[0106] In another broad form the present invention seeks to provide a method
for use in
assessing the likelihood of a biological subject having a medical condition,
the method
including, in one or more processing devices:
a) determining a pair of biomarker values, the pair of biomarker values being
selected from the group consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the PLA2G7 gene and PLAC8 gene;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide expression products of the CEACAM4 gene and LAMP] gene;
b) determining an indicator indicative of a ratio of the concentrations of the
polynucleotide expression products using the pair of biomarker values;
c) retrieving previously determined first and second indicator references from
a
database, the first and second indicator references being determined based on
indicators determined from first and second groups of a reference population,
one
of the groups consisting of individuals diagnosed with the medical condition;
d) comparing the indicator to the first and second indicator references;
e) using the results of the comparison to determine a probability indicative
of the
subject having the medical condition; and,
f) generating a representation of the probability, the representation being
displayed
to a user to allow the user to assess the likelihood of a biological subject
having at
least one medical condition.
[0107] Typically the method includes determining:
a) a first derived biomarker value indicative of a ratio of concentrations of
the
polynucleotide expression products using the first pair of biomarker values;
b) a second derived biomarker value indicative of a ratio of concentrations of
the
polynucleotide expression products using the first pair of biomarker values;
and,
c) determining the indicator by combining the first and second derived
biomarker
values.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 47 -
[0108] In another broad form the present invention seeks to provide apparatus
for
determining an indicator used in determining the likelihood of a biological
subject having at
least one medical condition, the apparatus including:
a) a sampling device that obtains a sample taken from a biological subject,
the
sample including polynucleotide expression products;
b) a measuring device that quantifies polynucleotide expression products
within the
sample to determine a pair of biomarker values, the pair of biomarker values
being selected from the group consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the PLA2G7 gene and PLAC8 gene;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide expression products of the CEACAM4 gene and LAMP] gene;
c) at least one processing device that:
i) receives an indication of the pair of biomarker values from the measuring
device;
ii) determines an indicator using a ratio of the concentration of the first
and
second polynucleotide expression products using the biomarker values; and,
iii) compares the indicator to at least one indicator reference; and,
iv) determines a likelihood of the subject having the at least one medical
condition using the results of the comparison; and,
v) generates a representation of the indicator and the likelihood for display
to a
user.
[0109] In another broad form the present invention seeks to provide a method
for
differentiating between inSIRS and ipSIRS in a biological subject, the method
including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) in a measuring device:
i) amplifying at least some polynucleotide expression products in the
sample;
ii) determining an amplification amount representing a degree of amplification

required to obtain a defined level of polynucleotide expression products
including:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 48 -
(1) amplification amounts for a first pair of polynucleotide expression
products of the PLA2G7 gene and PLAC8 gene;
(2) amplification amounts for a second pair of polynucleotide expression
products of the CEACAM4 gene and LAMP] gene;
c) in a processing system:
i) retrieving the amplification amounts;
ii) determining an indicator by:
(1) determining a first derived biomarker value indicative of a ratio of
concentrations of the first pair of polynucleotide expression products by
determining a difference between the amplification amounts for the first
pair;
(2) determining a second derived biomarker value indicative of a ratio of
concentrations of the second pair of polynucleotide expression products by
determining a difference between the amplification amounts for the second
pair;
(3) determining the indicator by adding the first and second derived biomarker

values;
iii) retrieving previously determined first and second indicator references
from a
database, wherein the first and second indicator references are distributions
of
indicators determined for first and second groups of a reference population,
the first and second group consisting of individuals diagnosed with inSIRS
and ipSIRS respectively;
iv) comparing the indicator to the first and second indicator references;
v) using the results of the comparison to determine a probability of the
subject
being classified within the first or second group;
vi) generating a representation at least partially indicative of the indicator
and the
probability; and,
vii)providing the representation to a user to allow the user to assess the
likelihood
of a biological subject having at least one medical condition.
[0110] In another broad form the present invention seeks to provide a method
for
determining an indicator used in assessing a likelihood of a biological
subject having a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 49 -
presence, absence, degree or prognosis of at least one medical condition, the
method
including:
a) determining a plurality of biomarker values, each biomarker value being
indicative of a value measured or derived for at least one corresponding
immune
system biomarker of the biological subject and being at least partially
indicative
of a concentration of the immune system biomarker in a sample taken from the
subject;
b) determining the indicator using a combination of the plurality of biomarker

values, wherein:
i) at least two biomarkers have a mutual correlation in respect of the at
least one
condition that lies within a mutual correlation range, the mutual correlation
range being between 0.9; and,
ii) the indicator has a performance value greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.
[0111] Typically the method includes:
c) determining a plurality of measured biomarker values, each measured
biomarker
value being a measured value of a corresponding biomarker of the biological
subject; and,
d) determining the indicator by applying a function to at least one of the
measured
biomarker values to determine at least one derived biomarker value, the at
least
one derived biomarker value being indicative of a value of a corresponding
derived biomarker.
[0112] Typically the function includes at least one of:
a) multiplying two biomarker values;
b) dividing two biomarker values;
c) adding two biomarker values;
d) subtracting two biomarker values;
e) a ratio of two biomarker values;
f) a weighted sum of at least two biomarker values;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 50 -
g) a log sum of at least two biomarker values; and,
h) a sigmoidal function of at least two biomarker values.
[0113] Typically the method includes determining at least one derived
biomarker value
corresponding to a ratio of two measured biomarker values.
[0114] Typically the method includes combining at least two biomarker values
to determine
an indicator value representing the indicator.
[0115] Typically the method includes combining at least two biomarker values
using a
combining function, the combining function being at least one of:
a) an additive model;
b) a linear model;
c) a support vector machine;
d) a neural network model;
e) a random forest model;
f) a regression model;
g) a genetic algorithm;
h) an annealing algorithm;
i) a weighted sum;
j) a nearest neighbor model; and,
k) a probabilistic model.
[0116] Typically at least one of the at least two biomarkers is a derived
biomarker.
[0117] Typically the method includes:
a) determining a first derived biomarker value, the first derived biomarker
value
being indicative of a ratio of concentrations of the first and second immune
system biomarkers;
b) determining a second derived biomarker value, the second derived biomarker
value being indicative of a ratio of concentrations of the third and fourth
measured
immune system biomarkers; and,
c) adding the first and second derived biomarker values to generate an
indicator
value.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-51 -
[0118] Typically the method is performed at least in part using an electronic
processing
device.
[0119] Typically the method includes, in the electronic processing device:
a) receiving a plurality of measured biomarker values, each measured biomarker

value being a measured value of a corresponding immune system biomarker;
b) applying a function to at least one of the measured biomarker values to
determine
at least one derived biomarker value, the at least one derived biomarker value
being indicative of a value of a corresponding derived biomarker; and,
c) combining at least one derived biomarker value and at least one other
biomarker
value to determine the indicator.
[0120] Typically the mutual correlation range is at least one of:
a) 0.8;
b) 0.7;
c) 0.6;
d) 0.5;
e) 0.4;
f) 0.3;
g) 0.2; and,
h) 0.1.
[0121] Typically each biomarker has a condition correlation with the presence,
absence,
degree or prognosis of the at least one condition that lies outside a
condition correlation
range, the condition correlation range being between 0.3.
[0122] Typically the condition correlation range is at least one of:
a) 0.9;
b) 0.8;
c) 0.7;
d) 0.6;
e) 0.5; and,
f) 0.4.
[0123] Typically the performance threshold is indicative of an explained
variance of at least
one of:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 52 -
a) 0.4;
b) 0.5;
c) 0.6;
d) 0.7;
e) 0.8; and,
f) 0.9.
[0124] Typically the biomarker value is indicative of a level or abundance of
a molecule
selected from one or more of a nucleic acid molecule and a proteinaceous
molecule.
[0125] Typically the method includes generating a representation of the
indicator.
[0126] Typically the representation includes:
a) an alphanumeric indication of the indicator;
b) a graphical indication of a comparison of the indicator to one or more
indicator
references;
c) an alphanumeric indication of a likelihood of the subject having at least
one
medical condition.
[0127] Typically the method includes:
a) comparing the indicator to an indicator reference; and,
b) determining a likelihood in accordance with results of the comparison.
[0128] Typically the indicator reference is based on at least one of:
a) an indicator threshold range;
b) an indicator threshold; and,
c) an indicator distribution.
[0129] Typically the indicator reference is derived from indicators determined
for a number
of individuals in a reference population.
[0130] Typically the indicator reference is based on a distribution of
indicators determined
for a group of a reference population, the group consisting of individuals
diagnosed with the
medical condition.
[0131] Typically the reference population includes:
a) a plurality of individuals of different sexes;
b) a plurality of individuals of different ethnicities;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 53 -
c) a plurality of healthy individuals;
d) a plurality of individuals suffering from at least one diagnosed medical
condition;
e) a plurality of individuals showing clinical signs of at least one medical
condition;
and,
f) first and second groups of individuals, each group of individuals
suffering from a
respective diagnosed medical condition.
[0132] Typically the indicator is for use in determining the likelihood that a
biological
subject has at least one medical condition, and wherein the reference
population includes:
a) individuals presenting with clinical signs of the at least one medical
condition;
b) individuals diagnosed with the at least one medical condition; and,
c) healthy individuals.
[0133] Typically the indicator reference is retrieved from a database.
[0134] Typically the likelihood is based on a probability generated using the
results of the
comparison.
[0135] Typically the indicator is for determining a likelihood of the subject
having a first or
second condition, and wherein the method includes:
a) comparing the indicator to first and second indicator references, the first
and
second indicator references being indicative of first and second conditions;
and,
b) determining the likelihood in accordance with the results of the
comparison.
[0136] Typically the method includes:
a) determining first and second indicator probabilities using the results of
the
comparisons; and,
b) combining the first and second indicator probabilities to determine a
condition
probability indicative of the likelihood.
[0137] Typically the first and second indicator references are distributions
of indicators
determined for first and second groups of a reference population, the first
and second group
consisting of individuals diagnosed with the first or second condition
respectively.
[0138] Typically the method includes:
a) obtaining a sample taken from the biological subject, the sample including
polynucleotide expression products;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 54 -
b) quantifying at least some of the polynucleotide expression products within
the
sample to determine at least a pair of biomarker values;
c) determining the indicator at least in part using the pair of biomarker
values;
[0139] Typically the method includes, determining the indicator at least in
part using a ratio
of concentrations of the polynucleotide expression products.
[0140] Typically the method includes:
a) quantifying polynucleotide expression products by:
b) amplifying at least some polynucleotide expression products in the sample;
and,
c) determining an amplification amount representing a degree of amplification
required to obtain a defined level of each of a pair of polynucleotide
expression
products; and,
d) determining the indicator by determining a difference between the
amplification
amounts.
[0141] Typically the amplification amount is at least one of:
a) a cycle time;
b) a number of cycles;
c) a cycle threshold;
d) an amplification time; and,
e) relative to an amplification amount of another amplified product.
[0142] Typically the method includes determining:
a) a first derived biomarker value by determining a difference between the
amplification amounts of a first pair of polynucleotide expression products;
b) a second derived biomarker value by determining a difference between the
amplification amounts of a second pair of polynucleotide expression products;
c) determining the indicator by adding the first and second derived biomarker
values.
[0143] Typically the immune system biomarker is an IRS biomarker of an immune
system of
the biological subject that is altered, or whose level of expression is
altered, as part of an
inflammatory response to damage or pathogenic insult.
[0144] Typically the indicator is for determining a likelihood of the subject
having at least
one of inSIRS and ipSIRS, and wherein the method includes:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 55 -
a) determining a first pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the PLA2G7 gene and PLAC8 gene;
b) determining a second pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the CEACAM4 gene and LAMP] gene; and,
c) determining the indicator using the first and second pairs of biomarker
values.
[0145] Typically the indicator is for determining a likelihood of the subject
having inSIRS or
ipSIRS, and wherein the method includes:
a) determining a first pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the PLA2G7 gene and PLAC8 gene;
b) determining a second pair of biomarker values indicative of a concentration
of
polynucleotide expression products of the CEACAM4 gene and LAMP] gene; and,
c) determining the indicator using the first and second pairs of biomarker
values.
[0146] Typically the indicator is for determining a likelihood of the subject
having inSIRS or
a healthy condition, and wherein biomarker values are determined from at least
one IRS
immune system biomarker in each of first and second IRS immune system
biomarker groups,
wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group A IRS immune system biomarker
genes; and
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group B IRS immune system biomarker
genes.
[0147] Typically the indicator is for determining a likelihood of the subject
having ipSIRS or
a healthy condition, and wherein biomarker values are determined from at least
one IRS
immune system biomarker in each of first and second IRS immune system
biomarker groups,
wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group C IRS immune system biomarker
genes; and,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 56 -
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group D IRS immune system biomarker
genes.
[0148] Typically the indicator is for determining a likelihood of the subject
having inSIRS or
ipSIRS, and wherein biomarker values are determined from at least one IRS
immune system
biomarker in each of first and second IRS immune system biomarker groups,
wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group E IRS immune system biomarker
genes; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group F IRS immune system biomarker
genes.
[0149] Typically the indicator is for determining a likelihood of the subject
having inSIRS or
ipSIRS, and wherein biomarker values are determined from at least one IRS
immune system
biomarker in each of first, second, third and fourth IRS immune system
biomarker groups,
wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group G IRS immune system biomarker
genes;
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group H IRS immune system biomarker
genes;
c) the third IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group I IRS immune system biomarker
genes; and,
d) the fourth IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group J IRS immune system biomarker
genes.
[0150] Typically the first IRS immune system biomarker is a PLA2G7 expression
product,
the second IRS immune system biomarker is a PLAC8 expression product, the
third IRS

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 57 -
immune system biomarker is a CEACAM4 expression product and the fourth IRS
immune
system biomarker is a LAMP] expression product.
[0151] Typically the indicator is for determining a likelihood of the subject
having mild
sepsis or severe sepsis, and wherein biomarker values are determined from at
least one IRS
immune system biomarker in each of first and second IRS immune system
biomarker groups,
wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group K IRS immune system biomarker
genes; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group L IRS immune system biomarker
genes.
[0152] Typically the indicator is for determining a likelihood of the subject
having mild
sepsis or septic shock, and wherein biomarker values are determined from at
least one IRS
immune system biomarker in each of first and second IRS immune system
biomarker groups,
wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group M IRS immune system biomarker
genes; and,
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group N IRS immune system biomarker
genes.
[0153] Typically the indicator is for determining a likelihood of the subject
having severe
sepsis or septic shock, and wherein biomarker values are determined from at
least one IRS
immune system biomarker in each of first and second IRS immune system
biomarker groups,
wherein:
a) the first IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group 0 IRS immune system biomarker
genes; and,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 58 -
b) the second IRS immune system biomarker group consists of polynucleotide
and/or
polypeptide expression products from group P IRS immune system biomarker
genes.
[0154] In another broad form the present invention seeks to provide apparatus
for
determining an indicator used in assessing a likelihood of a biological
subject having a
presence, absence, degree or prognosis of at least one medical condition, the
apparatus
including a processing device that:
a) determines a plurality of biomarker values, each biomarker value being
indicative
of a value measured or derived for at least one corresponding immune system
biomarker of the biological subject and being at least partially indicative of
a
concentration of the immune system biomarker in a sample taken from the
subject;
b) determines the indicator using a combination of the plurality of biomarker
values,
wherein:
i) at least two biomarkers have a mutual correlation in respect of the at
least one
condition that lies within a mutual correlation range, the mutual correlation
range being between 0.9; and,
ii) the indicator has a performance value greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.
[0155] In one broad form the present invention seeks to provide a method for
determining an
indicator for use in diagnosing the presence, absence, degree or prognosis of
at least one
condition in a biological subject, the method including:
a) determining a plurality of biomarker values, each biomarker value being
indicative of a value measured or derived for at least one corresponding
biomarker
of the biological subject;
b) determining the indicator using a combination of the plurality of biomarker

values, the at least one indicator being at least partially indicative of the
presence,
absence, degree or prognosis of the at least one condition, wherein:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 59 -
i) at least two markers have a mutual correlation in respect of the at least
one
condition that lies within a mutual correlation range, the mutual correlation
range being between 0.9; and,
ii) the indicator has a performance value greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.
[0156] Typically the method includes:
a) determining a plurality of measured biomarker values, each measured
biomarker
value being a measured value of a corresponding biomarker of the biological
subject; and,
b) applying a function to at least one of the measured biomarker values to
determine
at least one derived biomarker value, the at least one derived biomarker value

being indicative of a value of a corresponding derived biomarker.
[0157] Typically the function includes at least one of:
a) multiplying two biomarker values;
b) dividing two biomarker values;
c) adding two biomarker values;
d) subtracting two biomarker values;
e) a weighted sum of at least two biomarker values;
f) a log sum of at least two biomarker values; and,
g) a sigmoidal function of at least two biomarker values.
[0158] Typically the method includes determining at least one derived
biomarker value
corresponding to a ratio of two measured biomarker values.
[0159] Typically the method includes combining at least two biomarker values
to determine
an indicator value representing the indicator.
[0160] Typically the method includes combining at least two biomarker values
using a
combining function, the combining function being at least one of:
a) an additive model;
b) a linear model;
c) a support vector machine;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 60 -
d) a neural network model;
e) a random forest model;
f) a regression model;
g) a genetic algorithm;
h) an annealing algorithm;
i) a weighted sum;
j) a nearest neighbor model; and,
k) a probabilistic model.
[0161] Typically at least one of the at least two biomarkers is a derived
biomarker.
[0162] Typically the method includes:
a) determining a first derived biomarker value, the first derived biomarker
value
being a ratio of first and second measured biomarker values;
b) determining a second derived biomarker value, the second derived biomarker
value being a ratio of third and fourth measured biomarker values; and,
c) adding the first and second derived biomarker values to generate an
indicator
value.
[0163] Typically the method includes:
a) determining an indicator value;
b) comparing the indicator value to at least one indicator value range; and,
c) using a result of the comparison in diagnosing the presence, absence,
degree or
prognosis of at least one condition.
[0164] Typically the method is performed at least in part using an electronic
processing
device.
[0165] Typically the method includes, in the electronic processing device:
a) receiving a plurality of measured biomarker values, each measured biomarker

value being a measured value of a corresponding biomarker of the biological
subject;
b) applying a function to at least one of the measured biomarker values to
determine
at least one derived biomarker value, the at least one derived biomarker value
being indicative of a value of a corresponding derived biomarker; and,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 61 -
c) combining at least one derived biomarker value and at least one other
biomarker
value to determine an indicator value.
[0166] Typically the method includes generating a representation in accordance
with the at
least one indicator value.
[0167] Typically the method includes:
a) comparing the indicator value to at least one indicator value range; and,
b) displaying a result of the comparison.
[0168] Typically the mutual correlation range is at least one of:
a) 0.8;
b) 0.7;
c) 0.6;
d) 0.5;
e) 0.4;
f) 0.3;
g) 0.2; and,
h) 0.1.
[0169] Typically each biomarker has a condition correlation with the presence,
absence,
degree or prognosis of the at least one condition that lies outside a
condition correlation
range, the condition correlation range being between 0.3.
[0170] Typically the condition correlation range is at least one of:
a) 0.9;
b) 0.8;
c) 0.7;
d) 0.6;
e) 0.5; and,
f) 0.4.
[0171] Typically the performance threshold is indicative of an explained
variance of at least
one of:
a) 0.4;
b) 0.5;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 62 -
c) 0.6;
d) 0.7;
e) 0.8; and,
f) 0.9.
[0172] Typically the biomarker value is indicative of a level or abundance of
a molecule or
entity selected from one or more of:
a) A nucleic acid molecule;
b) A proteinaceous molecule;
c) An amino acid
d) A carbohydrate;
e) A lipid;
f) A steroid;
g) An inorganic molecule;
h) Anion;
i) A drug;
j) A chemical;
k) A metabolite;
1) A toxin;
m) A nutrient;
n) A gas;
o) A cell;
p) A pathogenic organism; and,
q) A non-pathogenic organism.
[0173] In another broad form the present invention seeks to provide apparatus
for
determining an indicator for use in diagnosing the presence, absence, degree
or prognosis of
at least one condition in a biological subject, the apparatus including an
electronic processing
device that:
a) determines a plurality of biomarker values, each biomarker value being
indicative
of a value measured or derived for at least one corresponding biomarker of the

biological subject;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 63 -
b) determines the indicator using a combination of the plurality of biomarker
values,
the at least one indicator being at least partially indicative of the
presence,
absence, degree or prognosis of the at least one condition, wherein:
i) at least two biomarkers have a mutual correlation in respect of the at
least one
condition that lies within a mutual correlation range, the mutual correlation
range being between 0.9; and,
ii) the indicator has a performance value greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.
[0174] In another broad form the present invention seeks to provide a
diagnostic signature
for use in diagnosing the presence, absence, degree or prognosis of at least
one condition in a
biological subject, the diagnostic signature defining a combination of at
least two biomarker
values corresponding to values of biomarkers that can be measured for or
derived from the
biological subject, wherein:
a) at least two biomarkers have a mutual correlation for the at least one
condition
that lies within a mutual correlation range, the mutual correlation range
being
between 0.9; and,
b) the combination of at least two biomarker values has a performance value
greater
than or equal to a performance threshold representing the ability of the
combination of the at least two biomarkers to diagnose the presence, absence,
degree or prognosis of the at least one condition, the performance threshold
being
a variance explained of at least 0.3.
[0175] Typically the diagnostic signature defines a function to be applied to
at least one
measured biomarker value to determine at least one derived biomarker value,
the at least one
derived biomarker value being indicative of a value of a corresponding derived
biomarker.
[0176] Typically the function includes at least one of:
a) multiplying two biomarker values;
b) dividing two biomarker values;
c) adding two biomarker values;
d) subtracting two biomarker values;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 64 -
e) a weighted sum of at least two biomarker values;
f) a log sum of at least two biomarker values; and,
g) a sigmoidal function of at least two biomarker values.
[0177] Typically the at least one derived biomarker value corresponds to a
ratio of two
measured biomarker values.
[0178] Typically the diagnostic signature defines a combination of at least
two biomarker
values for determining an indicator value representing the indicator.
[0179] Typically the diagnostic signature defines a combining function for
combining at least
two biomarker values, the combining function being at least one of:
a) an additive model;
b) a linear model;
c) a support vector machine;
d) a neural network model;
e) a random forest model;
f) a regression model;
g) a genetic algorithm;
h) an annealing algorithm; and,
i) a weighted sum.
[0180] Typically at least one of the at least two biomarkers is a derived
biomarker.
[0181] Typically the diagnostic signature defines:
a) a first derived biomarker value, the first derived biomarker value being a
ratio of
first and second measured biomarker values;
b) a second derived biomarker value, the second derived biomarker value being
a
ratio of third and fourth measured biomarker values; and,
c) a combination of the first and second derived biomarker values to generate
an
indicator value.
[0182] Typically the diagnostic signature defines at least one indicator value
range and
wherein comparison of at least one indicator value to the at least one
indicator value range is
used in diagnosing the presence, absence, degree or prognosis of at least one
condition.
[0183] Typically the mutual correlation range is at least one of:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 65 -
a) 0.8;
b) 0.7;
c) 0.6;
d) 0.5;
e) 0.4;
f) 0.3;
g) 0.2; and,
h) 0.1.
[0184] Typically each biomarker has a condition correlation with the presence,
absence,
degree or prognosis of the at least one condition that lies outside a
condition correlation
range, the condition correlation range being between 0.3.
[0185] Typically the condition correlation range is at least one of:
a) 0.9;
b) 0.8;
c) 0.7;
d) 0.6;
e) 0.5; and,
f) 0.4.
[0186] Typically the performance threshold is indicative of an explained
variance of at least
one of:
a) 0.4;
b) 0.5;
c) 0.6;
d) 0.7;
e) 0.8; and,
f) 0.9.
[0187] Typically the biomarker value is indicative of a level or abundance of
a molecule or
entity selected from one or more of:
a) A nucleic acid molecule;
b) A proteinaceous molecule;
c) An amino acid

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 66 -
d) A carbohydrate;
e) A lipid;
f) A steroid;
g) An inorganic molecule;
h) Anion;
i) A drug;
j) A chemical;
k) A metabolite;
1) A toxin;
m) A nutrient;
n) A gas;
o) A cell;
p) A pathogenic organism; and,
q) A non-pathogenic organism.
[0188] In another broad form the present invention seeks to provide a method
of identifying
biomarkers for use in a diagnostic signature, the diagnostic signature being
for use in
diagnosing the presence, absence, degree or prognosis of at least one
condition in a biological
subject, the method including:
a) for a number of candidate biomarkers, ranking the candidate biomarkers in
accordance with the ability of each biomarker to distinguish between the
presence,
absence, degree or prognosis of at least one condition in a biological
subject;
b) selecting at least two candidate biomarkers in accordance with the ranking,
the at
least two biomarkers having a mutual correlation for the at least one
condition that
lies within a mutual correlation range, the mutual correlation range being
between
0.9;
c) determining a performance value of a combination of the at least two
candidate
biomarkers; and,
d) defining a diagnostic signature in accordance with the combination of the
at least
two biomarkers if the performance value is greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 67 -
[0189] Typically the method includes determining a combination of at least two
candidate
biomarkers using a combining function, the combining function being at least
one of:
a) an additive model;
b) a linear model;
c) a support vector machine;
d) a neural network model;
e) a random forest model;
f) a regression model;
g) a genetic algorithm;
h) an annealing algorithm; and,
i) a weighted sum.
[0190] Typically the method includes:
a) selecting a next combining function;
b) determining if a performance value of a combination of the at least two
candidate
biomarkers determined by the next combining function is greater than or equal
to
a performance threshold; and,
c) if the performance value is not greater than or equal to a performance
threshold,
repeating steps a) and b) for successive combining functions.
[0191] Typically the method includes:
a) selecting two candidate biomarkers;
b) determining if a performance value of a combination of the two candidate
biomarkers is greater than or equal to a performance threshold; and,
c) if the performance value is not greater than or equal to a performance
threshold:
i) combining the selected candidate biomarkers with at least one additional
candidate biomarker; and,
ii) repeating steps a) and b) with at least one additional candidate
biomarker.
[0192] Typically the method includes combining a number of candidate
biomarkers up to a
limit.
[0193] Typically the method includes:
a) selecting a highest ranked candidate biomarker;
b) selecting a next highest ranked candidate biomarker;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 68 -
c) for the selected candidate markers, determining if the mutual correlation
for the
candidate biomarkers within the mutual correlation range; and,
d) if not, repeating steps b) and c) until two candidate biomarkers are
selected having
a mutual correlation within the mutual correlation range.
[0194] Typically the method includes:
a) defining at least two groups of candidate biomarkers, candidate biomarkers
in
different groups having a mutual correlation within the mutual correlation
range;
b) ranking the candidate biomarkers in each group; and,
c) selecting candidate biomarkers from the different groups.
[0195] Typically the method includes:
a) using reference data for at least one individual to define a number of
groups
indicative of the presence, absence, degree or prognosis of the at least one
condition; and,
b) using at least one analysis technique to identify a number of candidate
biomarkers
that are potentially useful for distinguishing the groups.
[0196] Typically the method includes using reference values measured for
reference
biomarkers for the at least one individual to identify the candidate
biomarkers.
[0197] Typically the method includes using reference values to filter
reference biomarkers to
determine candidate biomarkers.
[0198] Typically at least one of the candidate biomarkers is a derived
biomarker derived
from at least one of the reference biomarkers using a function.
[0199] Typically the derived biomarkers are derived from filtered biomarkers.
[0200] Typically the method includes:
a) applying a function to at least one of the reference values to determine at
least one
derived reference biomarker value, the at least one derived reference
biomarker
value being indicative of a value of a corresponding derived reference
biomarker;
and,
b) determining at least one candidate biomarker using the at least one derived

reference biomarker value.
[0201] Typically the method includes:

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 69 -
a) using reference data for at least one individual to define a number of
groups
indicative of the presence, absence, degree or prognosis of the at least one
condition; and,
b) for each group, combining a range of at least two reference biomarker
values to
determine an indicator value range for the group.
[0202] Typically the mutual correlation range is at least one of:
a) 0.8;
b) 0.7;
c) 0.6;
d) 0.5;
e) 0.4;
f) 0.3;
g) 0.2; and,
h) 0.1.
[0203] Typically each biomarker has a condition correlation with the presence,
absence,
degree or prognosis of the at least one condition that lies outside a
condition correlation
range, the condition correlation range being between 0.3.
[0204] Typically the condition correlation range is at least one of:
a) 0.9;
b) 0.8;
c) 0.7;
d) 0.6;
e) 0.5; and,
f) 0.4.
[0205] Typically the performance threshold is indicative of an explained
variance of at least
one of:
a) 0.4;
b) 0.5;
c) 0.6;
d) 0.7;
e) 0.8; and,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 70 -
f) 0.9.
[0206] In another broad form the present invention seeks to provide apparatus
for identifying
markers for use in a diagnostic signature, the diagnostic signature being for
use in diagnosing
the presence, absence, degree or prognosis of at least one condition in a
biological subject,
the apparatus including an electronic process device that:
a) for a number of candidate biomarkers, ranks the candidate biomarkers in
accordance with the ability of each biomarker to distinguish between the
presence,
absence, degree or prognosis of at least one condition in a biological
subject;
b) selects at least two candidate biomarkers in accordance with the ranking,
the at
least two biomarkers having a mutual correlation for the at least one
condition that
lies within a mutual correlation range, the mutual correlation range being
between
0.9;
c) determines a performance value of a combination of the at least two
candidate
biomarkers; and,
d) defines a diagnostic signature in accordance with the combination of the at
least
two biomarkers if the performance value is greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.
[0207] In another broad form the present invention seeks to provide a method
for diagnosing
the presence or absence of inSIRS or a healthy condition in a biological
subject, the method
comprising: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value
being indicative of a value measured or derived for at least one IRS biomarker
of a biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from inSIRS and a
healthy
condition, wherein: (i) at least two IRS biomarkers have a mutual correlation
in respect of the
at least one condition that lies within a mutual correlation range, the mutual
correlation range
being between 0.9; and (ii) the indicator has a performance value greater
than or equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 71 -
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group A IRS
biomarker genes
and wherein the second IRS biomarker group consists of polynucleotide and/or
polypeptide
expression products from group B IRS biomarker genes.
[0208] In another broad form the present invention seeks to provide a method
for diagnosing
the presence or absence of ipSIRS or a healthy condition in a biological
subject, the method
comprising: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value
being indicative of a value measured or derived for at least one IRS biomarker
of a biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from ipSIRS and a
healthy
condition, wherein: (i) at least two IRS biomarkers have a mutual correlation
in respect of the
at least one condition that lies within a mutual correlation range, the mutual
correlation range
being between 0.9; and (ii) the indicator has a performance value greater
than or equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group C IRS
biomarker genes
and wherein the second IRS biomarker group consists of polynucleotide and/or
polypeptide
expression products from group D IRS biomarker genes.
[0209] In another broad form the present invention seeks to provide a method
for diagnosing
the presence or absence of inSIRS or ipSIRS in a biological subject, the
method comprising:
(a) determining a plurality of IRS biomarker values, each IRS biomarker value
being
indicative of a value measured or derived for at least one IRS biomarker of a
biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 72 -
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from inSIRS and
ipSIRS, wherein:
(i) at least two IRS biomarkers have a mutual correlation in respect of the at
least one
condition that lies within a mutual correlation range, the mutual correlation
range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group E IRS
biomarker genes
and wherein the second IRS biomarker group consists of polynucleotide and/or
polypeptide
expression products from group F IRS biomarker genes.
[0210] In another broad form the present invention seeks to provide a method
for diagnosing
the presence or absence of inSIRS or ipSIRS in a biological subject, the
method comprising:
(a) determining a plurality of IRS biomarker values, each IRS biomarker value
being
indicative of a value measured or derived for at least one IRS biomarker of a
biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from inSIRS and
ipSIRS, wherein:
(i) at least four IRS biomarkers have a mutual correlation in respect of the
at least one
condition that lies within a mutual correlation range, the mutual correlation
range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least four IRS biomarkers is selected from a
first IRS biomarker
group, wherein at least one other of the at least four IRS biomarkers is
selected from a second
IRS biomarker group, wherein at least one other of the at least four IRS
biomarkers is
selected from a third IRS biomarker group, and wherein at least one other of
the at least four
IRS biomarkers is selected from a fourth IRS biomarker group, wherein the
first IRS

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 73 -
biomarker group consists of polynucleotide and/or polypeptide expression
products from
group G IRS biomarker genes, wherein the second IRS biomarker group consists
of
polynucleotide and/or polypeptide expression products from group H IRS
biomarker genes,
wherein the third IRS biomarker group consists of polynucleotide and/or
polypeptide
expression products from group I IRS biomarker genes and wherein the fourth
IRS biomarker
group consists of polynucleotide and/or polypeptide expression products from
group J IRS
biomarker genes.
[0211] Suitably, the first IRS biomarker is a PLA2G7 expression product, the
second IRS
biomarker is a PLAC8 expression product, the third IRS biomarker is a CEACAM4
expression product and the fourth IRS biomarker is a LAMP] expression product.
[0212] In another broad form the present invention seeks to provide a method
for diagnosing
the presence or absence of mild sepsis or severe sepsis in a biological
subject, the method
comprising: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value
being indicative of a value measured or derived for at least one IRS biomarker
of a biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from mild sepsis
and severe sepsis,
wherein: (i) at least two IRS biomarkers have a mutual correlation in respect
of the at least
one condition that lies within a mutual correlation range, the mutual
correlation range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group K IRS
biomarker genes
and wherein the second IRS biomarker group consists of polynucleotide and/or
polypeptide
expression products from group L IRS biomarker genes.
[0213] In another form the present invention seeks to provide a method for
diagnosing the
presence or absence of mild sepsis or septic shock in a biological subject,
the method

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 74 -
comprising: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value
being indicative of a value measured or derived for at least one IRS biomarker
of a biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from mild sepsis
and septic shock,
wherein: (i) at least two IRS biomarkers have a mutual correlation in respect
of the at least
one condition that lies within a mutual correlation range, the mutual
correlation range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group M IRS
biomarker genes
and wherein the second IRS biomarker group consists of polynucleotide and/or
polypeptide
expression products from group N IRS biomarker genes.
[0214] In another form the present invention seeks to provide a method for
diagnosing the
presence or absence of severe sepsis or septic shock in a biological subject,
the method
comprising: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value
being indicative of a value measured or derived for at least one IRS biomarker
of a biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from severe sepsis
and septic shock,
wherein: (i) at least two IRS biomarkers have a mutual correlation in respect
of the at least
one condition that lies within a mutual correlation range, the mutual
correlation range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 75 -
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group 0 IRS
biomarker genes
and wherein the second IRS biomarker group consists of polynucleotide and/or
polypeptide
expression products from group P IRS biomarker genes.
[0215] In another broad form the present invention seeks to provide a kit
comprising: (i) a
reagent that allows quantification of a first IRS biomarker; and (ii) a
reagent that allows
quantification of a second IRS biomarker, wherein the first and second IRS
biomarkers have
a mutual correlation in respect of at least one condition selected from a
healthy condition,
ipSIRS or a stage of ipSIRS selected from mild sepsis, severe sepsis and
septic
shock, which at least one condition lies within a mutual correlation range of
between 0.9,
and wherein a combination of respective biomarker values for the first and
second IRS
biomarkers that are measured for or derived from a biological subject has a
performance
value greater than or equal to a performance threshold representing the
ability of the
combination of the first and second IRS biomarkers to diagnose the presence,
absence or
degree of the at least one condition, or to provide a prognosis for the at
least one condition,
the performance threshold being a variance explained of at least 0.3.
[0216] Suitably, the kit further comprises: (iii) a reagent that allows
quantification of a third
IRS biomarker; and (iv) a reagent that allows quantification of a fourth IRS
biomarker,
wherein the third and fourth IRS biomarkers have a mutual correlation in
respect of at the
least one condition that lies within a mutual correlation range of between
0.9, and wherein a
combination of respective biomarker values for the third and fourth IRS
biomarkers that are
measured for or derived from a biological subject has a performance value
greater than or
equal to a performance threshold representing the ability of the combination
of the third and
fourth IRS biomarkers to diagnose the presence, absence or degree of the at
least one
condition, or to provide a prognosis for the at least one condition, the
performance threshold
being a variance explained of at least 0.3.
[0217] Suitably, the kit is for diagnosing the presence or absence of inSIRS
or a healthy
condition, wherein the first IRS biomarker is selected from a first IRS
biomarker group and
wherein the second IRS biomarker is selected from a second IRS biomarker
group, wherein
the first IRS biomarker group consists of polynucleotide and/or polypeptide
expression
products from group A IRS biomarker genes and wherein the second IRS biomarker
group

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 76 -
consists of polynucleotide and/or polypeptide expression products from group B
IRS
biomarker genes.
[0218] Suitably, the kit is for diagnosing the presence or absence of ipSIRS
or a healthy
condition, wherein the first IRS biomarker is selected from a first IRS
biomarker group and
wherein the second IRS biomarker is selected from a second IRS biomarker
group, wherein
the first IRS biomarker group consists of polynucleotide and/or polypeptide
expression
products from group C IRS biomarker genes and wherein the second IRS biomarker
group
consists of polynucleotide and/or polypeptide expression products from group D
IRS
biomarker genes.
[0219] Suitably, the kit is for diagnosing the presence or absence of inSIRS
or ipSIRS,
wherein the first IRS biomarker is selected from a first IRS biomarker group
and wherein the
second IRS biomarker is selected from a second IRS biomarker group, wherein
the first IRS
biomarker group consists of polynucleotide and/or polypeptide expression
products from
group E IRS biomarker genes and wherein the second IRS biomarker group
consists of
polynucleotide and/or polypeptide expression products from group F IRS
biomarker genes.
[0220] Suitably, the kit is for diagnosing the presence or absence of inSIRS
or ipSIRS,
wherein the first IRS biomarker is selected from a first IRS biomarker group,
wherein the
second IRS biomarker is selected from a second IRS biomarker group, wherein
the third IRS
biomarker is selected from a third IRS biomarker group and wherein the fourth
IRS
biomarker is selected from a fourth IRS biomarker group, wherein the first IRS
biomarker
group consists of polynucleotide and/or polypeptide expression products from
group G IRS
biomarker genes, wherein the second IRS biomarker group consists of
polynucleotide and/or
polypeptide expression products from group H IRS biomarker genes, wherein the
third IRS
biomarker group consists of polynucleotide and/or polypeptide expression
products from
group I IRS biomarker genes and wherein the fourth IRS biomarker group
consists of
polynucleotide and/or polypeptide expression products from group J IRS
biomarker genes.
[0221] Suitably, the first IRS biomarker is a PLA2G7 expression product, the
second IRS
biomarker is a PLAC8 expression product, the third IRS biomarker is a CEACAM4
expression product and the fourth IRS biomarker is a LAMP] expression product.
[0222] Suitably, the kit is for diagnosing the presence or absence of mild
sepsis or severe
sepsis, wherein the first IRS biomarker is selected from a first IRS biomarker
group and

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 77 -
wherein the second IRS biomarker is selected from a second IRS biomarker
group, wherein
the first IRS biomarker group consists of polynucleotide and/or polypeptide
expression
products from group K IRS biomarker genes and wherein the second IRS biomarker
group
consists of polynucleotide and/or polypeptide expression products from group L
IRS
biomarker genes.
[0223] Suitably, the kit is for diagnosing the presence or absence of mild
sepsis or septic
shock, wherein the first IRS biomarker is selected from a first IRS biomarker
group and
wherein the second IRS biomarker is selected from a second IRS biomarker
group, wherein
the first IRS biomarker group consists of polynucleotide and/or polypeptide
expression
products from group M IRS biomarker genes and wherein the second IRS biomarker
group
consists of polynucleotide and/or polypeptide expression products from group N
IRS
biomarker genes.
[0224] Suitably, the kit is for diagnosing the presence or absence of severe
sepsis or septic
shock, wherein the first IRS biomarker is selected from a first IRS biomarker
group and
wherein the second IRS biomarker is selected from a second IRS biomarker
group, wherein
the first IRS biomarker group consists of polynucleotide and/or polypeptide
expression
products from group 0 IRS biomarker genes and wherein the second IRS biomarker
group
consists of polynucleotide and/or polypeptide expression products from group P
IRS
biomarker genes.
[0225] In another broad form the present invention seeks to provide a method
for treating,
preventing or inhibiting the development of at least one condition selected
from inSIRS,
ipSIRS or a particular stage of ipSIRS (e.g., mild sepsis, severe sepsis or
septic shock) in a
subject, the method comprising (a) determining a plurality of IRS biomarker
values, each IRS
biomarker value being indicative of a value measured or derived for at least
one IRS
biomarker of a biological subject; (b) determining an indicator using a
combination of the
plurality of IRS biomarker values, the indicator being at least partially
indicative of the
presence, absence or degree of the at least one condition, wherein: (i) at
least two IRS
biomarkers have a mutual correlation in respect of the at least one condition
that lies within a
mutual correlation range, the mutual correlation range being between 0.9; and
(ii) the
indicator has a performance value greater than or equal to a performance
threshold
representing the ability of the indicator to diagnose the presence, absence or
degree of the at

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 78 -
least one condition, the performance threshold being indicative of an
explained variance of at
least 0.3; and (c) administering to the subject, on the basis that the
indicator indicates the
presence of inSIRS, an effective amount of an agent that treats or ameliorates
the symptoms
or reverses or inhibits the development of inSIRS, or administering to the
subject, on the
basis that the indicator indicates the presence of ipSIRS or a particular
stage of ipSIRS, an
effective amount of an agent that treats or ameliorates the symptoms or
reverses or inhibits
the development of ipSIRS or the particular stage of ipSIRS.
[0226] Suitably, the method further comprises: (1) determining a plurality of
measured IRS
biomarker values, each measured IRS biomarker value being a measured value of
an IRS
biomarker of the biological subject; and (2) applying a function to at least
one of the
measured IRS biomarker values to determine at least one derived IRS biomarker
value, the at
least one derived IRS biomarker value being indicative of a value of a
corresponding derived
IRS biomarker.
[0227] Suitably, the function includes at least one of: (a) multiplying two
IRS biomarker
values; (b) dividing two IRS biomarker values; (c) adding two IRS biomarker
values; (d)
subtracting two IRS biomarker values; (e) a weighted sum of at least two IRS
biomarker
values; (f) a log sum of at least two IRS biomarker values; and (g) a
sigmoidal function of at
least two IRS biomarker values.
[0228] In another broad form the present invention seeks to provide a method
of monitoring
the efficacy of a particular treatment regimen in a subject towards a desired
health state, the
method comprising: a) determining a plurality of IRS biomarker values, each
IRS biomarker
value being indicative of a value measured or derived for at least one IRS
biomarker of a
biological subject after treatment with a treatment regimen; (b) determining
an indicator
using a combination of the plurality of IRS biomarker values, the indicator
being at least
partially indicative of the presence, absence or degree of at least one
condition selected from
a healthy condition, inSIRS, ipSIRS or a particular stage of ipSIRS, wherein:
(i) at least two
IRS biomarkers have a mutual correlation in respect of the at least one
condition that lies
within a mutual correlation range, the mutual correlation range being between
0.9; and (ii)
the indicator has a performance value greater than or equal to a performance
threshold
representing the ability of the indicator to diagnose the presence, absence or
degree of the at
least one condition, or to provide a prognosis for the at least one condition,
the performance

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 79 -
threshold being indicative of an explained variance of at least 0.3, and (c)
determining that
the treatment regimen is effective for changing the health status of the
subject to the desired
health state on the basis that the indicator indicates the presence of a
healthy condition or the
presence of a condition of a lower degree relative to the degree of the
condition in the subject
before treatment with the treatment regimen.
[0229] In another broad form the present invention seeks to provide a method
of correlating a
biomarker signature with an effective treatment regimen for a condition
selected from
inSIRS, ipSIRS or a particular stage of ipSIRS (e.g., mild sepsis, severe
sepsis and septic
shock), the method comprising: (a) determining a biomarker signature defining
a
combination of at least two IRS biomarker values corresponding to values of at
least two IRS
biomarkers that can be measured for or derived from a biological subject
having the
condition and for whom an effective treatment has been identified, wherein:
(i) the at least
two IRS biomarkers have a mutual correlation in respect of the condition that
lies within a
mutual correlation range, the mutual correlation range being between 0.9; and
(ii) the
combination of at least two biomarker values has a performance value greater
than or equal
to a performance threshold representing the ability of the combination of at
least two
biomarker values to diagnose the presence, absence or degree of the condition,
or to provide
a prognosis for the condition, the performance threshold being indicative of
an explained
variance of at least 0.3; and (b) correlating the biomarker signature so
determined with an
effective treatment regimen for the condition.
[0230] In another broad form the present invention seeks to provide a method
of determining
whether a treatment regimen is effective for treating a subject with a
condition selected from
inSIRS, ipSIRS or a particular stage of ipSIRS (e.g., mild sepsis, severe
sepsis and septic
shock), the method comprising: (a) determining a plurality of post-treatment
IRS biomarker
values, each post-treatment IRS biomarker value being indicative of a value
measured or
derived for at least one IRS biomarker of a biological subject after treatment
with the
treatment regimen; (b) determining a post-treatment indicator using a
combination of the
plurality of post-treatment IRS biomarker values, the post-treatment indicator
being at least
partially indicative of the presence, absence or degree of at least one
condition selected from
a healthy condition, inSIRS, ipSIRS or a particular stage of ipSIRS, wherein:
(i) at the least
two IRS biomarkers have a mutual correlation in respect of the at least one
condition that lies

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 80 -
within a mutual correlation range, the mutual correlation range being between
0.9; and (ii)
the post-treatment indicator has a performance value greater than or equal to
a performance
threshold representing the ability of the post-treatment indicator to diagnose
the presence,
absence or degree of the at least one condition, the performance threshold
being indicative of
an explained variance of at least 0.3, wherein the post-treatment indicator
indicates whether
the treatment regimen is effective for treating the condition in the subject
on the basis that
post-treatment indicator indicates the presence of a healthy condition or the
presence of a
condition of a lower degree relative to the degree of the condition in the
subject before
treatment with the treatment regimen.
[0231] In another broad form the present invention seeks to provide a method
of correlating a
biomarker signature with a positive or negative response or a side effect to a
treatment
regimen, the method comprising: (a) determining a biomarker signature defining
a
combination of at least two IRS biomarker values corresponding to values of at
least two IRS
biomarkers that can be measured for or derived from a biological subject
following
commencement of the treatment regimen, wherein: (i) the at least two IRS
biomarkers have a
mutual correlation in respect of at least one condition selected from a
healthy condition,
inSIRS, ipSIRS or a particular stage of ipSIRS (e.g., mild sepsis, severe
sepsis and septic
shock), which lies within a mutual correlation range, the mutual correlation
range being
between 0.9; and (ii) the combination of at least two biomarker values has a
performance
value greater than or equal to a performance threshold representing the
ability of the
combination of at least two biomarker values to diagnose the presence, absence
or degree of
the at least one condition, or to provide a prognosis for the at least one
condition, the
performance threshold being indicative of an explained variance of at least
0.3; and (b)
correlating the biomarker signature so determined with a positive or negative
response to the
treatment regimen.
[0232] In another broad form the present invention seeks to provide a method
of determining
a positive or negative response to a treatment regimen and/or a side effect of
a treatment
regimen by a subject with a condition selected from inSIRS, ipSIRS or a
particular stage of
ipSIRS (e.g., mild sepsis, severe sepsis and septic shock), the method: (a)
correlating a
reference biomarker signature with a positive or negative response or a side
effect to the
treatment regimen, wherein the biomarker signature defines a combination of at
least two IRS

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 81 -
biomarker values corresponding to values of at least two IRS biomarkers that
are measured
for or derived from a control biological subject or control group, wherein:
(i) the at least two
IRS biomarkers have a mutual correlation in respect of at least one condition
selected from a
healthy condition, inSIRS, ipSIRS or a particular stage of ipSIRS (e.g., mild
sepsis, severe
sepsis and septic shock), which lies within a mutual correlation range, the
mutual correlation
range being between 0.9; and (ii) the combination of at least two biomarker
values has a
performance value greater than or equal to a performance threshold
representing the ability of
the combination of at least two biomarker values to diagnose the presence,
absence or degree
of the at least one condition, or to provide a prognosis for the at least one
condition, the
performance threshold being indicative of an explained variance of at least
0.3; (b)
determining a sample biomarker signature defining a combination of at least
two IRS
biomarker values corresponding to values of at least two IRS biomarkers that
are measured
for or derived from a biological subject following commencement of the
treatment regimen,
wherein: (i) the at least two IRS biomarkers have a mutual correlation in
respect of at least
one condition selected from a healthy condition, inSIRS, ipSIRS or a
particular stage of
ipSIRS, which lies within a mutual correlation range, the mutual correlation
range being
between 0.9; and (ii) the combination of at least two biomarker values has a
performance
value greater than or equal to a performance threshold representing the
ability of the
combination of at least two biomarker values to diagnose the presence, absence
or degree of
the at least one condition, or to provide a prognosis for the at least one
condition, the
performance threshold being indicative of an explained variance of at least
0.3; wherein the
sample biomarker signature indicates whether the subject is responding
positively or
negatively to the treatment regimen and/or is developing a side effect from
the treatment
regimen, based on the correlation of the reference biomarker signature with
the positive or
negative response or side effect to the treatment regimen.
[0233] In another broad form the present invention seeks to provide a method
of determining
a positive or negative response to a treatment regimen and/or a side effect to
a treatment
regimen by a biological subject, the method comprising: (a) determining a
sample biomarker
signature defining a combination of at least two IRS biomarker values
corresponding to
values of at least two IRS biomarkers that are measured for or derived from a
biological
subject following commencement of the treatment regimen, wherein: (i) the at
least two IRS
biomarkers have a mutual correlation in respect of at least one condition
selected from a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 82 -
healthy condition, inSIRS, ipSIRS or a particular stage of ipSIRS (e.g., mild
sepsis, severe
sepsis and septic shock), which lies within a mutual correlation range, the
mutual correlation
range being between 0.9; and (ii) the combination of at least two biomarker
values has a
performance value greater than or equal to a performance threshold
representing the ability of
the combination of at least two biomarker values to diagnose the presence,
absence or degree
of the at least one condition, or to provide a prognosis for the at least one
condition, the
performance threshold being indicative of an explained variance of at least
0.3, wherein the
sample biomarker signature is correlated with a positive or negative response
to the treatment
regimen and/or to a side effect from the treatment regimen; and (b)
determining whether the
subject is responding positively or negatively to the treatment regimen and/or
is developing a
side effect from the treatment regimen based on the sample biomarker
signature.
Brief Description of the Drawings
[0234] An example of the present invention will now be described with
reference to the
accompanying drawings, in which: -
[0235] Figure 1A is a flowchart of an example of a method for deriving an
indicator for use
in diagnosing the presence, absence or degree of at least one condition or in
providing a
prognosis of at least one condition in a biological subject;
[0236] Figure 1B is a flowchart of an example of a method for identifying
biomarkers for use
in a biomarker signature;
[0237] Figure 2 is a schematic diagram of an example of a distributed computer
architecture;
[0238] Figure 3 is a schematic diagram of an example of a processing system of
Figure 2;
[0239] Figure 4 is a schematic diagram of an example of a computer system of
Figure 2;
[0240] Figure 5 is a flowchart of a specific example of a method for
identifying biomarkers
for use in a biomarker signature;
[0241] Figure 6A is a flowchart of a first example of a method for selecting
candidate
biomarkers;
[0242] Figure 6B is a flowchart of a second example of a method for selecting
candidate
biomarkers;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 83 -
[0243] Figure 7 is a flowchart of a second example of a method for use in
diagnosing the
presence, absence or degree of at least one condition or in providing a
prognosis of at least
one condition in a biological subject;
[0244] Figure 8A is a plot of 941 mRNA biomarkers against the AUC for
differentiating
between healthy condition and post-surgical inflammation (PS) (also referred
to herein as
"infection-negative SIRS" (inSIRS)), for individual biomarkers having an AUC
greater than
0.7;
[0245] Figure 8B is a box and whisker plot showing the best mRNA biomarker for
separating
healthy condition and PS;
[0246] Figure 8C is a plot of the AUC for the diagnostic ability of 1000
derived biomarkers
in separating healthy condition and PS with all derived biomarkers having an
AUC of 1.0;
[0247] Figure 8D is a box and whisker plot of the best performing derived
biomarker, based
on AUC, for separating healthy condition and PS;
[0248] Figures 8E and 8F are two plots showing the correlation to each other
of the
biomarkers in each group;
[0249] Figures 8G and 8H are two plots demonstrating the AUC of the biomarkers
in each
group (groups 1 and 2);
[0250] Figure 81 is a box and whisker plot showing that when biomarkers are
derived from
group 1 and group 2 that a greater overall AUC is obtained;
[0251] Figure 9A is a plot of the 941 mRNA biomarkers against the AUC for
differentiating
between healthy condition and sepsis (also referred to herein as "infection-
positive SIRS"
(ipSIRS)) with all individual biomarkers having an AUC greater than 0.7;
[0252] Figure 9B is a box and whisker plot showing the best mRNA biomarker for
separating
healthy condition and sepsis;
[0253] Figure 9C is a plot of the AUC for the diagnostic ability of 1000
derived biomarkers
in separating healthy condition and sepsis with all derived biomarkers having
an AUC of 1.0;
[0254] Figure 9D is a box and whisker plot of the best performing derived
biomarker, based
on AUC, for separating healthy condition and sepsis;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 84 -
[0255] Figures 9E and 9F show the correlation to each other of the biomarkers
groups
correlated to each group;
[0256] Figures 9G and 9H are two plots demonstrating the AUC of the biomarkers
in each
group (bucketgroups 1 and 2);
[0257] Figure 91 is a box and whisker plot showing that when biomarkers are
derived from
bucketgroup 1 and bucketgroup 2 that a greater overall AUC is obtained;
[0258] Figure 10A is a plot of the 359 mRNA biomarkers against the AUC for
differentiating
between PS and sepsis with all individual biomarkers having had an AUC greater
than 0.7;
[0259] Figure 10B is a box and whisker plot showing the best mRNA biomarker
for
separating PS and sepsis;
[0260] Figure 10C is a plot of the AUC for the diagnostic ability of 1000
derived biomarkers
in separating PS and sepsis with all derived biomarkers having an AUC of 0.9;
[0261] Figure 10D is a box and whisker plot of the best derived biomarker for
separating PS
and sepsis;
[0262] Figures 10E is a plot showing the correlation of the biomarkers in each
bucketgroup
to the condition;
[0263] Figures 1OF is a box and whisker plot showing that when biomarkers are
derived from
bucketgroup 1 and bucketgroup 2 that a greater overall AUC is obtained;
[0264] Figures 10G is a plot demonstrating the AUC of the biomarkers in each
of four
bucketgroups;
[0265] Figure 10H is a box and whisker plot showing that when biomarkers are
derived from
each of the four bucketgroups a greater overall AUC is obtained;
[0266] Figure 11A is a plot of the 66 mRNA biomarkers against the AUC for
differentiating
between mild sepsis and severe sepsis with all individual biomarkers selected
having an AUC
greater than 0.7;
[0267] Figure 11B is a box and whisker plot showing the best mRNA biomarker
for
separating mild sepsis and severe sepsis;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 85 -
[0268] Figure 11C is a plot of the AUC for the diagnostic ability of 1000
derived biomarkers
in separating mild sepsis and severe sepsis with all derived biomarkers have
an AUC of at
least 0.87;
[0269] Figure 11D is a box and whisker plot of the best performing derived
biomarker, based
on AUC, for separating mild sepsis and severe sepsis;
[0270] Figures 11E and 11F are plots showing the correlation to each other of
the biomarkers
in each group;
[0271] Figures 11G and 11H are plots demonstrating the AUC of the biomarkers
in each
group;
[0272] Figure 111 is a box and whisker plot showing that when biomarkers are
derived from
group 1 and group 2 that a greater overall AUC is obtained;
[0273] Figure 12A is a plot of the 48 mRNA biomarkers against the AUC for
differentiating
between mild sepsis and septic shock (also referred to herein as "infection-
positive SIRS-
shock" (ipSIRS-shock)) with all individual biomarkers having an AUC greater
than 0.7;
[0274] Figure 12B is a box and whisker plot showing the best mRNA biomarker
for
separating mild sepsis and septic shock;
[0275] Figure 12C is a plot of the AUC for the diagnostic ability of 1000
derived biomarkers
in separating mild sepsis and septic shock with all derived biomarkers having
an AUC of at
least 0.793;
[0276] Figure 12D is a box and whisker plot of the best performing derived
biomarker, based
on AUC, for separating mild sepsis and septic shock;
[0277] Figures 12E and 12F are plots showing the correlation to each other of
the biomarkers
in each group;
[0278] Figures 12G and 12H are plots demonstrating the AUC of the biomarkers
in each
group;
[0279] Figure 121 is a box and whisker plot showing that when biomarkers are
derived from
group 1 and group 2 that a greater overall AUC is obtained;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 86 -
[0280] Figure 13A is a plot of the 61 mRNA biomarkers against the AUC for
differentiating
between severe sepsis and septic shock with all individual biomarkers selected
having an
AUC greater than 0.7;
[0281] Figure 13B is a box and whisker plot showing the best mRNA biomarker
for
separating severe sepsis and septic shock;
[0282] Figure 13C is a plot of the AUC for the diagnostic ability of 1000
derived biomarkers
in separating severe sepsis and septic shock with all derived biomarkers have
an AUC of at
least 0.821;
[0283] Figure 13D is a box and whisker plot of the best performing derived
biomarker, based
on AUC, for separating severe sepsis and septic shock;
[0284] Figures 13E and 13F are plots showing the correlation to each other of
the biomarkers
in each group;
[0285] Figures 13G and 13H are plots demonstrating the AUC of the biomarkers
in each
group;
[0286] Figure 131 is a box and whisker plot showing that when biomarkers are
derived from
group 1 and group 2 that a greater overall AUC is obtained;
[0287] Figure 14 is a user interface illustrating an example of a thermal
cycler protocol;
[0288] Figure 15 is a diagram of an example of a report; and,
[0289] Figures 16A to 16L are box and whisker plots for the top twelve
biomarker
combinations for distinguishing between healthy condition and PS;
[0290] Figures 17A to 17L are box and whisker plots for the top twelve
biomarker
combinations for distinguishing between healthy condition and sepsis;
[0291] Figures 18A to 18L are box and whisker plots for the top twelve
biomarker
combinations for distinguishing between PS and sepsis;
[0292] Figures 19A to 19L are box and whisker plots for the top twelve
biomarker
combinations for distinguishing between sepsis and severe sepsis;
[0293] Figures 20A to 20L are box and whisker plots for the top twelve
biomarker
combinations for distinguishing between severe sepsis and septic shock;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 87 -
[0294] Figures 21A to 21L are box and whisker plots for the top twelve
biomarker
combinations for distinguishing between sepsis and septic shock;
[0295] Figure 22 is a graph of the effect on AUC of adding biomarkers to a
biomarker
signature;
[0296] Figure 23 is a graph showing an example of the ability of the biomarker
signature to
distinguish between PS and sepsis for two patient populations;
[0297] Figure 24 is a flowchart of an example of a method for determining
indicator
references;
[0298] Figures 25A and 25B are a flowchart of an example of a method for
validating an
indicator derived from biomarker measurements;
[0299] Figures 26 is an example showing the comparison of an indicator value
to an indicator
reference; and,
[0300] Figures 27A and 27B are example representation of indicator values.
Detailed Description of the Preferred Embodiments
[0301] An example of a method for determining an indicator for use in
diagnosing the
presence, absence or degree of at least one condition in or of a biological
subject, or in
monitoring the progression of at least one condition in or of the subject, or
in prognosing at
least one condition in or of the subject, will now be described with reference
to Figure 1A.
[0302] For the purpose of explanation, a number of different terms will be
used. For
example, the term "biomarker" refers to a measurable parameter, or combination
of
parameters, that can be used as an indicator of a biological state and
includes, without
limitation, proteins, nucleic acids, carbohydrates, lipids, metabolites,
gases, steroids, ions,
nutrients, toxins, cells, pathogenic organisms, non-pathogenic organisms,
organic compounds
and inorganic compounds. Biomarkers also encompass non-blood-borne factors,
non-analyte
physiological markers of health status, or other factors or biomarkers not
measured from
samples (e.g., biological samples such as bodily fluids), such as "clinical"
or "phenotypic"
parameters, including, without limitation, age, ethnicity, gender, species,
breed, genetic
information, white blood cell count, diastolic blood pressure and systolic
blood pressure,
bone density, height, weight, waist and hip circumference, body-mass index, as
well as others

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 88 -
such as Type I or Type II diabetes mellitus or gestational diabetes mellitus
(collectively
referred to here as diabetes), resting heart rate, homeostatic model
assessment (HOMA),
HOMA insulin resistance (HOMA-IR), intravenous glucose tolerance (SI(IVGT)),
resting
heart rate, f3 cell function, macrovascular function, microvascular function,
atherogenic
index, low-density lipoprotein/high-density lipoprotein ratio, intima-media
thickness, body
temperature, sequential organ failure assessment (SOFA) and the like. The
"biomarkers"
could also include "immune response biomarkers", which will be described in
more detail
below.
[0303] The term "biomarker value" refers to a value measured or derived for at
least one
corresponding biomarker of the biological subject and which is typically at
least partially
indicative of a concentration of the immune system biomarker in a sample taken
from the
subject. Thus, the biomarker values could be measured biomarker values, which
are values
of biomarkers measured for the subject, or alternatively could be derived
biomarker values,
which are values that have been derived from one or more measured biomarker
values, for
example by applying a function to the one or more measured biomarker values.
[0304] Biomarker values can be of any appropriate form depending on the manner
in which
the values are determined. For example, the biomarker values could be
determined using
high-throughput technologies such as mass spectrometry, sequencing platforms,
array and
hybridization platforms, immunoassays, flow cytometry, or any combination of
such
technologies and in one preferred example, the biomarker values relate to a
level of activity
or abundance of an expression product or other measurable molecule, quantified
using a
technique such as PCR, sequencing or the like. In this case, the biomarker
values can be in
the form of amplification amounts, or cycle times, which are a logarithmic
representation of
the concentration of the biomarker within a sample, as will be appreciated by
persons skilled
in the art and as will be described in more detail below.
[0305] The term "reference biomarkers" is used to refer to biomarkers whose
activity has
been quantified for a sample population of one or more individuals having one
or more
conditions, stages of one or more conditions, subtypes of one or more
conditions or different
prognoses. The term "reference data" refers to data measured for one or more
individuals in a
sample population, and may include quantification of the level or activity of
the biomarkers

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 89 -
measured for each individual, information regarding any conditions of the
individuals, and
optionally any other information of interest.
[0306] The term "candidate biomarkers" refers to a subset of the reference
biomarkers that
have been identified as being potentially useful in distinguishing between
different groups of
individuals, such as individuals suffering from different conditions, or
having different stages
or prognoses. The number of candidate biomarkers will vary, but is typically
about 200.
[0307] The term "signature biomarkers" is used to refer to a subset of the
candidate
biomarkers that have been identified for use in a biomarker signature that can
be used in
performing a clinical assessment, such as to rule in or rule out a specific
condition, different
stages or severity of conditions, subtypes of different conditions or
different prognoses. The
number of signature biomarkers will vary, but is typically of the order of 10
or less.
[0308] The term "biomarker signature" means a combination of at least two
biomarker
values corresponding to values of biomarkers that can be measured for or
derived from one
or more biological subjects, which combination is characteristic for a
discrete condition,
stage of condition, subtype of condition or a prognosis for a discrete
condition, stage of
condition, subtype of condition.
[0309] The terms "biological subject", "subject", "individual" and "patient"
are used
interchangeably herein to refer to an animal subject, particularly a
vertebrate subject, and
even more particularly a mammalian subject. Suitable vertebrate animals that
fall within the
scope of the invention include, but are not restricted to, any member of the
phylum Chordata,
subphylum vertebrata including primates, rodents (e.g., mice rats, guinea
pigs), lagomorphs
(e.g., rabbits, hares), bovines (e.g., cattle), ovines (e.g., sheep), caprines
(e.g., goats), porcines
(e.g., pigs), equines (e.g., horses), canines (e.g., dogs), felines (e.g.,
cats), avians (e.g.,
chickens, turkeys, ducks, geese, companion birds such as canaries, budgerigars
etc.), marine
mammals (e.g., dolphins, whales), reptiles (snakes, frogs, lizards, etc.), and
fish. A preferred
subject is a primate (e.g., a human, ape, monkey, chimpanzee).
[0310] As used herein, the term SIRS ("systemic inflammatory response
syndrome") refers
to a clinical response arising from a non-specific insult with two or more of
the following
measureable clinical characteristics; a body temperature greater than 38 C or
less than 36
C, a heart rate greater than 90 beats per minute, a respiratory rate greater
than 20 per minute,
a white blood cell count (total leukocytes) greater than 12,000 per mm3 or
less than 4,000 per

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 90 -
mm3, or a band neutrophil percentage greater than 10%. From an immunological
perspective,
it may be seen as representing a systemic response to insult (e.g., major
surgery) or systemic
inflammation. As used herein, "inSIRS" (which includes within its scope "post-
surgical"
(PS) inflammation) includes the clinical response noted above but in the
absence of a
systemic infectious process. By contrast, "ipSIRS" (also referred to herein as
"sepsis")
includes the clinical response noted above but in the presence of a presumed
or confirmed
infection. Confirmation of infection can be determined using microbiological
culture or
isolation of the infectious agent. From an immunological perspective, ipSIRS
may be seen as
a systemic response to microorganisms, whether it is a local, peripheral or
systemic infection.
[0311] As used herein, the term "degree" of a condition refers to the
seriousness, severity,
stage or state of a condition. For example, a condition may be characterized
as mild,
moderate or severe. A person of skill in the art would be able to determine or
assess the
degree of a particular condition. For example, the degree of a condition may
be determined
by comparing the likelihood or length of survival of a subject having a
condition with the
likelihood or length of survival in other subjects having the same condition.
In other
embodiments, the degree of a condition may be determined by comparing the
clinical signs
of a subject having a condition with the degree of the clinical signs in other
subjects having
the same condition.
[0312] It will be appreciated that the above described terms and associated
definitions are
used for the purpose of explanation only and are not intended to be limiting.
[0313] In this example, the method includes determining a plurality of
biomarker values at
step 100, each biomarker value being indicative of a value measured or derived
for at least
one biomarker of the biological subject.
[0314] The biomarker values and biomarkers corresponding to the biomarker
values can be
of any appropriate form and in particular can relate to any attribute of a
subject for which a
value can be quantified. This technique is particularly suited to high-
throughput technologies
such as mass spectrometry, sequencing platforms, array and hybridization
platforms, or any
combination of such technologies and in one preferred example, the biomarker
values relate
to a level of activity or abundance of an expression product or other
measurable molecule.
[0315] The biomarker values could be measured biomarker values, which are
values of
biomarkers measured for the subject, or alternatively could be derived
biomarker values,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 91 -
which are values that have been derived from one or more measured biomarker
values, for
example by applying a function to the one or more measured biomarker values.
As used
herein, biomarkers to which a function has been applied are referred to as
"derived markers".
[0316] The biomarker values may be determined in any one of a number of ways.
In one
example, the process of determining the biomarker values can include measuring
the
biomarker values, for example by performing tests on the biological subject.
More typically
however, the step of determining the biomarker values includes having an
electronic
processing device receive or otherwise obtain biomarker values that have been
previously
measured or derived. This could include for example, retrieving the biomarker
values from a
data store such as a remote database, obtaining biomarker values that have
been manually
input, using an input device, or the like.
[0317] At step 110, the indicator is determined using a combination of the
plurality of
biomarker values, the indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition.
[0318] The biomarker values can be combined in any one of a number of ways and
this can
include for example adding, multiplying, subtracting, or dividing biomarker
values to
determine an indicator value. This step is performed so that multiple
biomarker values can
be combined into a single indicator value, providing a more useful and
straightforward
mechanism for allowing the indicator to be interpreted and hence used in
diagnosing the
presence, absence or degree of the at least one condition in the subject, or
prognosing the at
least one condition in the subject.
[0319] Assuming the method is performed using an electronic processing device,
at step 120
an indication of the indicator is optionally displayed or otherwise provided
to the user. In
this regard, the indication could be a graphical or alphanumeric
representation of an indicator
value. Alternatively however, the indication could be the result of a
comparison of the
indicator value to predefined thresholds or ranges, or alternatively could be
an indication of
the presence, absence, degree or prognosis for at least one condition, derived
using the
indicator.
[0320] In order to ensure an effective diagnosis or prognosis can be
determined, at least two
of the biomarkers have a mutual correlation in respect of the at least one
condition that lies
within a mutual correlation range, the mutual correlation range being between
0.9. This

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 92 -
requirement means that the two biomarkers are not entirely correlated in
respect of each other
when considered in the context of the condition(s) being diagnosed or
prognosed. In other
words, at least two of the biomarkers in the combination respond differently
as the condition
changes, which adds significantly to their ability when combined to
discriminate between at
least two conditions, to diagnose the presence, absence or degree of at least
one condition,
and/or to provide a prognosis of at least condition in or of the biological
subject. As used
herein, "and/or" refers to and encompasses any and all possible combinations
of one or more
of the associated listed items, as well as the lack of combinations when
interpreted in the
alternative (or).
[0321] Typically, the requirement that biomarkers have a low mutual
correlation means that
the biomarkers may relate to different biological attributes or domains such
as but not limited
to different molecular functions, different biological processes and different
cellular
components. Illustrative examples of molecular function include addition of or
removal of
one of more of the following moieties to or from a protein, polypeptide,
peptide, nucleic acid
(e.g., DNA, RNA): linear, branched, saturated or unsaturated alkyl (e.g.,
C1¨C24 alkyl);
phosphate; ubiquitin; acyl; fatty acid, lipid, phospholipid; nucleotide base;
hydroxyl and the
like. Molecular functions also include signaling pathways, including without
limitation,
receptor signaling pathways and nuclear signaling pathways. Non-limiting
examples of
molecular functions also include cleavage of a nucleic acid, peptide,
polypeptide or protein at
one or more sites; polymerization of a nucleic acid, peptide, polypeptide or
protein;
translocation through a cell membrane (e.g., outer cell membrane; nuclear
membrane);
translocation into or out of a cell organelle (e.g., Golgi apparatus,
lysosome, endoplasmic
reticulum, nucleus, mitochondria); receptor binding, receptor signaling,
membrane channel
binding, membrane channel influx or efflux; and the like.
[0322] Illustrative examples of biological processes include: stages of the
cell cycle such as
meiosis, mitosis, cell division, prophase, metaphase, anaphase, telophase and
interphase,
stages of cell differentiation; apoptosis; necrosis; chemotaxis; immune
responses including
adaptive and innate immune responses, pro-inflammatory immune responses,
autoimmune
responses, tolerogenic responses and the like. Other illustrative examples of
biological
processes include generating or breaking down adenosine triphosphate (ATP),
saccharides,
polysaccharides, fatty acids, lipids, phospholipids, sphingolipids,
glycolipids, cholesterol,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 93 -
nucleotides, nucleic acids, membranes (e.g., cell plasma membrane, nuclear
membrane),
amino acids, peptides, polypeptides, proteins and the like.
[0323] Representative examples of cellular components include organelles,
membranes, as
for example noted above, and others.
[0324] It will be appreciated that the use of biomarkers that have different
biological
attributes or domains provides further information than if the biomarkers were
related to the
same or common biological attributes or domains.
[0325] In this regard, it will be appreciated if the at least two biomarkers
are highly
correlated to each other, the use of both biomarkers would add little
diagnostic/prognostic
improvement compared to the use of a single one of the biomarkers.
Accordingly, the
method uses biomarkers that are not well correlated with each other, thereby
ensuring that the
inclusion of each biomarker in the method adds significantly to the
discriminative ability of
the indicator.
[0326] Despite this, in order to ensure that the indicator can accurately be
used in performing
the discrimination between at least two conditions or the diagnosis of the
presence, absence
or degree of at least one condition or the prognosis of at least one
condition, the indicator has
a performance value that is greater than or equal to a performance threshold.
The
performance threshold may be of any suitable form but is to be typically
indicative of an
explained variance of at least 0.3, or an equivalent value of another
performance measure.
[0327] It has been found that utilizing a combination of biomarkers that have
a mutual
correlation between 0.9 and using these in a combination that provides an
explained
variance of at least 0.3, this allows an indicator to be defined that is
suitable for ensuring that
an accurate discrimination, diagnosis or prognosis can be obtained whilst
minimizing the
number of biomarkers that are required.
[0328] It will be appreciated that in this context, the biomarkers used within
the above-
described method can define a biomarker signature for the at least one
condition, which
includes a minimal number of biomarkers, whilst maintaining sufficient
performance to allow
the biomarker signature to be used in making a clinically relevant diagnosis,
prognosis, or
differentiation. Minimizing the number of biomarkers used minimizes the costs
associated
with performing diagnostic or prognostic tests and in the case of nucleic acid
expression

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 94 -
products, allows the test to be performed utilizing relatively straightforward
techniques such
as nucleic acid array, and polymerase chain reaction (PCR) processes, or the
like, allowing
the test to be performed rapidly in a clinical environment.
[0329] Furthermore, producing a single indicator value allows the results of
the test to be
easily interpreted by a clinician or other medical practitioner, so that test
can be used for
reliable diagnosis in a clinical environment.
[0330] An example of the process for generating a suitable biomarker signature
for use in the
method of Figure 1A will now be described with reference to Figure 1B.
[0331] In particular, it is typical to generate a biomarker signature by
analyzing a large
number of biomarkers and then selecting a combination of biomarkers that meet
the above
described criteria.
[0332] In this example, at step 150 the process includes ranking a number of
candidate
biomarkers in accordance with the ability of each biomarker to distinguish
between the
presence, absence, degree or prognosis of at least one condition in a
biological subject.
[0333] The candidate biomarkers can be obtained in any appropriate matter, but
typically this
would involve acquiring reference data including reference biomarker values
relating to a
number of reference biomarkers that have been measured or derived for one or
more
reference individuals having different presences, absences, degrees or
prognoses of the one or
more conditions of interest. Thus, it will be appreciated that the candidate
biomarkers can
include measured and/or derived biomarkers, as will be described in more
detail below.
[0334] The reference data typically includes measurements of a plurality of
reference
biomarkers, the measurements including information regarding the activity,
such as the level,
abundance or functional activity, of any expression product or measurable
molecule, as will
be described in more detail below. The reference data may also include
information such as
clinical data regarding one or more conditions suffered by each individual.
This can include
information regarding a presence, absence, degree or progression of a
condition, phenotypic
information, such as details of phenotypic traits, genetic or genetically
regulated information,
amino acid or nucleotide related genomics information, results of other tests
including
imaging, biochemical and hematological assays, other physiological scores such
as a SOFA

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 95 -
(Sequential Organ Failure Assessment) score, or the like and this is not
intended to be
limiting, as will be apparent from the description below.
[0335] The candidate biomarkers can include some or all of the reference
biomarkers,
depending on the preferred implementation. Thus, for example, reference
biomarker values
could be analyzed to determine correlations between the reference biomarkers
and the at least
one condition, with the reference biomarkers being coarsely filtered to remove
those with a
low correlation, for example with a correlation with the condition that is
below 0.3.
[0336] At step 160 at least two candidate biomarkers are selected based on the
ranking and a
mutual correlation. In particular, at least two candidate biomarkers are
selected which have a
mutual correlation within a mutual correlation range of 0.9. Thus, this
process excludes any
biomarkers which are highly mutually correlated, when considered in the
context of the one
or more conditions, and which would not therefore add significantly to the
ability to
discriminate between the presence, absence, degree or prognosis of at least
one condition.
[0337] At step 170 a performance value of a combination of the selected
candidate
biomarkers is determined. As mentioned above the combination may be any
combination of
the candidate biomarker values, such as addition, subtraction, multiplication,
or division of
the candidate biomarker values, and this will not therefore be described in
any further detail.
[0338] At step 180 it is determined if the performance value of the
combination exceeds a
performance threshold, the performance threshold being equivalent to an
explained variance
of at least 0.3. If so, the combination of the candidate biomarkers can be
used to define a
biomarker signature. Otherwise, the previous steps can be repeated, for
example by
determining alternative combinations of the candidate biomarkers, selecting
different
candidate biomarkers, or adding additional candidate biomarkers as will be
described in more
detail below. In this regard, it will be appreciated that other measures could
be used, and
reference to an explained variance of at least 0.3 is intended to be a
particular example for
illustrative purposes.
[0339] Accordingly, the above described method can be utilized to select a
combination of
candidate biomarkers that are suitable for use as signature biomarkers in a
biomarker
signature for diagnosing the presence, absence, degree or prognosis of at
least one condition
in a biological subject, for example using the method of Figure 1A above.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 96 -
[0340] In one example, this is achieved by ensuring that at least two of the
biomarkers used
are not highly mutually correlated, thereby ensuring that each of these
biomarkers contributes
to the performance of the resulting signature.
[0341] A number of further features will now be described.
[0342] In one example, the method includes determining a plurality of measured
biomarker
values, each measured biomarker value being a measured value of a
corresponding biomarker
of the biological subject and applying a function to at least one of the
measured biomarker
values to determine at least one derived biomarker value, the at least one
derived biomarker
value being indicative of a value of a corresponding derived biomarker.
[0343] The function used will therefore vary depending on the preferred
implementation. In
one example, the function includes at least one of multiplying two biomarker
values; dividing
two biomarker values; adding two biomarker values; subtracting two biomarker
values; a
weighted sum of at least two biomarker values; a log sum of at least two
biomarker values;
and, a sigmoidal function of at least two biomarker values.
[0344] More typically the function is division of two biomarker values, so
that the derived
biomarker value corresponds to a ratio of two measured biomarker values. There
are a
number of reasons why the ratio might be preferred. For example, use of a
ratio is self-
normalizing, meaning variations in measuring techniques will automatically be
accommodated. For example, if the input concentration of a sample is doubled,
the relative
proportions of biomarkers will remain the same. As a result, the type of
function therefore
has a stable profile over a range of input concentrations, which is important
because input
concentration is a known variable for expression data. Additionally, many
biomarkers are
nodes on biochemical pathways, so the ratio of biomarkers gives information
about the
relative activation of one biological pathway to another, which is a natural
representation of
biological change within a system. Finally, ratios are typically easily
interpreted.
[0345] The method typically includes combining at least two biomarker values
to determine
an indicator value representing the indicator. This is usually achieved by
combining at least
two biomarker values using a combining function, such as: an additive model; a
linear model;
a support vector machine; a neural network model; a random forest model; a
regression
model; a genetic algorithm; an annealing algorithm; and a weighted sum.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 97 -
[0346] In one example, at least one of the at least two biomarkers is a
derived biomarker and
in a preferred example, the combining function is addition of derived
biomarker values that
are ratios, in which case the method includes determining first and second
derived biomarker
values from ratios of first and second and third and fourth measured biomarker
values, and
then adding the first and second derived biomarker values to generate an
indicator value.
[0347] In one example, the method includes determining an indicator value,
comparing the
indicator value to at least one indicator value range, and using a result of
the comparison in
diagnosing the presence, absence, degree or prognosis of at least one
condition.
[0348] The above-described process is typically performed using an electronic
processing
device, forming part of a processing system such as a computer system or the
like. In this
case, the method typically involves having the electronic processing device
receive a
plurality of measured biomarker values, apply a function to at least one of
the measured
biomarker values to determine the at least one derived biomarker value and
combining at
least one derived biomarker value and at least one other biomarker value to
determine an
indicator value.
[0349] The electronic processing device can then generate a representation in
accordance
with the at least one indicator value, for example by displaying a numerical
indication of the
indicator value. More typically however the electronic processing device
compares the
indicator value to at least one indicator value range and displays a result of
the comparison.
This can be used to compare the indicator to defined ranges representing
specific stages,
progressions or prognoses of one or more conditions, allowing an indication of
the respective
stage, progression or prognosis to be displayed.
[0350] The mutual correlation range is typically at least one of: 0.8; 0.7;
0.6; 0.5; 0.4;
0.3; 0.2; and 0.1. In this regard, it will be appreciated that the
smaller the mutual
correlation range used, the less correlated the biomarkers will be and hence
the more useful
these will be in discriminating between the specific stages, progressions or
prognoses of one
or more conditions.
[0351] It is also typical for the biomarkers used to have at least a minimum
correlation with
the condition. In one example, each biomarker has a condition correlation with
the presence,
absence, degree or prognosis of the at least one condition that lies outside a
condition
correlation range, the condition correlation range being between 0.3.
However, it will be

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 98 -
appreciated that the greater the range, the greater the correlation between
the biomarker and
condition, and hence the more use this will be in performing a diagnosis.
Accordingly, the
condition correlation is more typically one of: 0.9; 0.8; 0.7; 0.6; 0.5;
and 0.4.
[0352] Furthermore, it will be appreciated that the greater the performance of
the indicator,
the greater use the indicator will be in performing the diagnosis.
Accordingly, the
performance threshold is typically indicative of an explained variance of at
least one of: 0.4;
0.5; 0.6; 0.7; 0.8; and 0.9.
[0353] As described above, the biomarker value can be of any suitable form.
However, the
technique is particularly suited to biomarker values indicative of a level or
abundance of a
molecule selected from one or more of: a nucleic acid molecule; a
proteinaceous molecule;
an amino acid; a carbohydrate; a lipid; a steroid; an inorganic molecule; an
ion; a drug; a
chemical; a metabolite; a toxin; a nutrient; a gas; a cell; a pathogenic
organism; and a non-
pathogenic organism.
[0354] When determining biomarkers for use in a biomarker signature, the
method typically
includes selecting a combining function, determining if a performance value of
a
combination of the at least two candidate biomarkers determined by the
combining function
is greater than or equal to a performance threshold and if the performance
value is not greater
than or equal to a performance threshold, repeating these steps for successive
different
combining functions. Thus, this allows a number of different combining
functions to be tried
successively, allowing the best combining function to be identified.
[0355] The method typically further includes selecting two candidate
biomarkers,
determining if a performance value of a combination of the two candidate
biomarkers is
greater than or equal to a performance threshold and if not, combining the
selected candidate
biomarkers with at least one additional candidate biomarker before repeating
the steps with at
least one additional candidate biomarker. This allows a larger number of
biomarkers to be
used in the event two biomarkers are insufficient, and this can be repeated,
with increasing
numbers of candidate biomarkers used in combination and compared to the
performance
threshold until the required performance is reached, or up until a defined
number limit of
candidate biomarkers is reached.
[0356] To ensure the required mutual correlation is met when selecting the
candidate
biomarkers, the method typically includes selecting a highest and a next
highest ranked

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 99 -
candidate biomarker, for the selected candidate biomarkers, determining if the
mutual
correlation for the candidate biomarkers within the mutual correlation range
and if not
repeating these steps until two candidate biomarkers are selected having a
mutual correlation
within the mutual correlation range. Alternatively however this can be
achieved by defining
at least two groups of candidate biomarkers, candidate biomarkers in different
groups having
a mutual correlation within the mutual correlation range, ranking the
candidate biomarkers in
each group and selecting candidate biomarkers from the different groups.
[0357] In general, the candidate biomarkers are determined by using reference
data for at
least one individual to define a number of groups indicative of the presence,
absence, degree
or prognosis of the at least one condition and then using at least one
analysis technique to
identify a number of candidate biomarkers that are potentially useful for
distinguishing the
groups. The groups can also be used to establish a range of at least two
reference biomarker
values to determine an indicator value range for the group.
[0358] In one example, reference values measured for reference biomarkers for
the at least
one individual can then be used to identify the candidate biomarkers, for
example by filtering
reference biomarkers to determine candidate biomarkers based on a correlation
of each
biomarker with the condition.
[0359] In one example, the process is performed by one or more processing
systems
operating as part of a distributed architecture, an example of which will now
be described
with reference to Figure 2.
[0360] In this example, the arrangement includes a number of processing
systems 201 and
computer systems 203 interconnected via one or more communications networks,
such as the
Internet 202, and/or a number of local area networks (LANs) 204. It will be
appreciated that
the configuration of the networks 202, 204 is for the purpose of example only,
and in practice
the processing and computer systems 201, 203 can communicate via any
appropriate
mechanism, such as via wired or wireless connections, including, but not
limited to mobile
networks, private networks, such as an 802.11 networks, the Internet, LANs,
WANs, or the
like, as well as via direct or point-to-point connections, such as Bluetooth,
or the like.
[0361] The use of separate terms "processing system" and "computer system" is
for
illustrative purposes and to enable distinction between different devices,
optionally having
different functionality. For example, the processing and computer systems 201,
203 could

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 100 -
represent servers and clients respectively, as will become apparent from the
following
description. However, this is not intended to be limiting and in practice any
suitable
computer network architecture can be used.
[0362] An example of a suitable processing system 201 is shown in Figure 3. In
this
example, the processing system 201 includes an electronic processing device,
such as at least
one microprocessor 300, a memory 301, an optional input/output device 302,
such as a
keyboard and/or display, and an external interface 303, interconnected via a
bus 304 as
shown. In this example the external interface 303 can be utilized for
connecting the
processing system 201 to peripheral devices, such as the communications
networks 202, 204,
databases 211, other storage devices, or the like. Although a single external
interface 303 is
shown, this is for the purpose of example only, and in practice multiple
interfaces using
various methods (e.g., Ethernet, serial, USB, wireless or the like) may be
provided.
[0363] In use, the microprocessor 300 executes instructions in the form of
applications
software stored in the memory 301 to perform required processes, such as
communicating
with other processing or computer systems 201, 203. Thus, actions performed by
a
processing system 201 are performed by the processor 300 in accordance with
instructions
stored as applications software in the memory 301 and/or input commands
received via the
I/0 device 302, or commands received from other processing or computer systems
201, 203.
The applications software may include one or more software modules, and may be
executed
in a suitable execution environment, such as an operating system environment,
or the like.
[0364] Accordingly, it will be appreciated that the processing systems 201 may
be formed
from any suitable processing system, such as a suitably programmed computer
system, PC,
web server, network server, or the like. In one particular example, the
processing systems 201
are standard processing system such as a 32-bit or 64-bit Intel Architecture
based processing
system, which executes software applications stored on non-volatile (e.g.,
hard disk) storage,
although this is not essential. However, it will also be understood that the
processing system
could be or could include any electronic processing device such as a
microprocessor,
microchip processor, logic gate configuration, firmware optionally associated
with
implementing logic such as an FPGA (Field Programmable Gate Array), or any
other
electronic device, system or arrangement.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 101 -
[0365] As shown in Figure 4, in one example, the computer systems 203 include
an
electronic processing device, such as at least one microprocessor 400, a
memory 401, an
input/output device 402, such as a keyboard and/or display, and an external
interface 403,
interconnected via a bus 404 as shown. In this example the external interface
403 can be
utilized for connecting the computer system 203 to peripheral devices, such as
the
communications networks 202, 204, databases, other storage devices, or the
like. Although a
single external interface 403 is shown, this is for the purpose of example
only, and in practice
multiple interfaces using various methods (e.g., Ethernet, serial, USB,
wireless or the like)
may be provided.
[0366] In use, the microprocessor 400 executes instructions in the form of
applications
software stored in the memory 401 to perform required processes, for example
to allow
communication with other processing or computer systems 201, 203. Thus,
actions
performed by a processing system 203 are performed by the processor 400 in
accordance
with instructions stored as applications software in the memory 401 and/or
input commands
received from a user via the I/0 device 402. The applications software may
include one or
more software modules, and may be executed in a suitable execution
environment, such as an
operating system environment, or the like.
[0367] Accordingly, it will be appreciated that the computer systems 203 may
be formed
from any suitable processing system, such as a suitably programmed PC,
Internet terminal,
lap-top, hand-held PC, smart phone, PDA, tablet, or the like. Thus, in one
example, the
processing system 300 is a standard processing system such as a 32-bit or 64-
bit Intel
Architecture based processing system, which executes software applications
stored on non-
volatile (e.g., hard disk) storage, although this is not essential. However,
it will also be
understood that the processing systems 203 can be any electronic processing
device such as a
microprocessor, microchip processor, logic gate configuration, firmware
optionally
associated with implementing logic such as an FPGA (Field Programmable Gate
Array), or
any other electronic device, system or arrangement.
[0368] It will also be noted that whilst the processing and computer systems
201, 203 are
shown as single entities, it will be appreciated that this is not essential,
and instead one or
more of the processing and/or computer systems 201, 203 can be distributed
over

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 102 -
geographically separate locations, for example by using processing systems
provided as part
of a cloud based environment.
[0369] Examples of the above-described method(s) will now be described in
further detail.
For the purpose of these examples, it is assumed that the process is performed
by one or more
of the processing systems 201, acting as diagnostic servers. Interaction by a
user is via a user
computer system 203, which is used to allow a user to submit raw data, for
example obtained
from measurements on a subject, with the processing system 201 generating the
indicator,
allowing this to be displayed on the computer system 203.
[0370] However, it will be appreciated that the above-described configuration
assumed for
the purpose of the following examples is not essential, and numerous other
configurations
may be used. It will also be appreciated that the partitioning of
functionality between the
processing and computer systems 201, 203 may vary, depending on the particular

implementation.
[0371] An example of a specific method for identifying biomarkers for using a
biomarker
signature will now be described with reference to Figure 5.
[0372] In this example, at step 500 reference data is obtained from at least
one individual.
The reference data is typically in the form of measured biomarker values
obtained for at least
one individual for different stages of the at least one condition.
[0373] The reference data may be acquired in any appropriate manner but
typically this
involves obtaining gene expression product data from a plurality of
individuals, selected to
include individuals diagnosed with one or more conditions of interest, as well
as healthy
individuals. Detection of either types of gene expression in use of any of the
methods
described herein is encompassed by the present invention. The terms
"expression" or "gene
expression" refer to production of RNA only or production of RNA and
translation of RNA
into proteins or polypeptides. Thus, the term "expression products" encompass
(i)
polynucleotides including RNA transcripts and corresponding nucleic acids
including
complementary cDNA copies of RNA transcripts, and (ii) polypeptides encoded by
RNA
transcripts. The conditions are typically medical, veterinary or other health
status conditions
and may include any illness, disease, stages of disease, disease subtypes,
severities of disease,
diseases of varying prognoses or the like. The terms "healthy individual",
"healthy subject"
and the like are used herein to refer to a subject, in particular a mammal,
having no diagnosed

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 103 -
disease, disorder, infirmity, or ailment. The condition of such an individual
or subject is
referred to herein as a "healthy condition" such that in one example a
condition can include
healthy. In specific embodiments, a healthy subject lacks SIRS (e.g., inSIRS
or ipSIRS).
[0374] In order to achieve this, gene expression product data are collected,
for example by
obtaining a biological sample, such as a peripheral blood sample, and then
performing a
quantification process, such as a nucleic acid amplification process,
including PCR
(Polymerase Chain Reaction) or the like, in order to assess the activity, and
in particular,
level or abundance of a number of reference biomarkers. Quantified values
indicative of the
relative activity are then stored as part of the reference data.
[0375] Example reference biomarkers could include expression products such as
nucleic acid
or proteinaceous molecules, as well as other molecules relevant in making a
clinical
assessment. The number of biomarkers measured for use as reference biomarkers
will vary
depending upon the preferred implementation, but typically include a large
number such as,
1000, 5000, 10000 or above, although this is not intended to be limiting.
[0376] The individuals also typically undergo a clinical assessment allowing
any conditions
to be clinically identified, and with an indication of any assessment or
condition forming part
of the reference data. Whilst any conditions can be assessed, in one example
the process is
utilized specifically to identify conditions such as SIRS (Systemic
Inflammatory Response
Syndrome) (M S Rangel-Frausto, D Pittet, M Costigan, T Hwang, C S Davis, and R
P
Wenzel, "The Natural History of the Systemic Inflammatory Response Syndrome
(SIRS). a
Prospective Study.", JAMA : the Journal of the American Medical Association
273, no. 2
(January 11, 1995): 117-123.). SIRS is an overwhelming whole body reaction
that may have
an infectious or non-infectious etiology, whereas sepsis is SIRS that occurs
during infection.
Both are defined by a number of non-specific host response parameters
including changes in
heart and respiratory rate, body temperature and white cell counts (Mitchell M
Levy et al.,
"2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference",
Critical
Care Medicine 31, no. 4 (April 2003): 1250-1256.; K Reinhart, M Bauer, N C
Riedemann,
and C S Hartog, "New Approaches to Sepsis: Molecular Diagnostics and
Biomarkers",
Clinical Microbiology Reviews 25, no. 4 (October 3,2012): 609-634) To
differentiate these
conditions they are referred herein to as SIRS (both conditions), infecti Oil-
negative SIRS
(SIRS without infection, hereafter referred to as "inSIRS") and infection-
positive SIRS

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 104 -
(sepsis, SIRS with a known or suspected infection, hereafter referred to as
"ipSIRS"). The
causes of SIRS are multiple and varied and can include, but are not limited
to, trauma, burns,
pancreatitis, endotoxemia, surgery, adverse drug reactions, and infections
(local and
systemic). It will be appreciated from the following, however, that this can
be applied to a
range of different conditions, and reference to SIRS or sepsis is not intended
to be limiting.
[0377] Additionally, the reference data may include additional biomarkers such
as one or
more phenotypic or clinical parameters of the individuals and/or their
relatives. Phenotypic
parameters can include information such as the gender, ethnicity, age, hair
color, eye color,
height, weight, waist and hip circumference, or the like. Also, in the case of
the technology
being applied to individuals other than humans, this can also include
information such as
designation of a species, breed or the like. Clinical traits may include
genetic information,
white blood cell count, diastolic blood pressure and systolic blood pressure,
bone density,
body-mass index, diabetes, resting heart rate, HOMA, HOMAIR, IVGT, resting
heart rate,
cell function, macrovascular function, microvascular function, atherogenic
index, low-density
lipoprotein/high-density lipoprotein ratio, intima-media thickness, body
temperature, SOFA
and the like.
[0378] Accordingly, in one example the reference data can include for each of
the reference
individuals information relating to at least one and desirably to a plurality
of reference
biomarkers and a presence, absence, degree or progression of a condition, .
[0379] The reference data may be collected from individuals presenting at a
medical center
with clinical signs relating to relevant any conditions of interest, and may
involve follow-on
consultations in order to confirm clinical assessments, as well as to identify
changes in
biomarkers, and/or clinical signs, and/or severity of clinical signs, over a
period of time. In
this latter case, the reference data can include time series data indicative
of the progression of
a condition, and/or the activity of the reference biomarkers, so that the
reference data for an
individual can be used to determine if the condition of the individual is
improving, worsening
or static. It will also be appreciated that the reference biomarkers are
preferably substantially
similar for the individuals within the sample population, so that comparisons
of measured
activities between individuals can be made.
[0380] This reference data could also be collected from a single individual
over time, for
example as a condition within the individual progresses, although more
typically it would be

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 105 -
obtained from multiple individuals each of which has a different stage of the
one or more
conditions of interest.
[0381] It will be appreciated that once collected, the reference data can be
stored in the
database 211 allowing this to be subsequently retrieved by the processing
system 201 for
subsequent analysis. The processing system 201 also typically stores an
indication of an
identity of each of the reference biomarkers.
[0382] In one example, the measurements are received as raw data, which then
undergoes
preliminary processing. Such raw data corresponds to information that has come
from a
source without modification, such as outputs from instruments such as PCR
machines, array
(e.g., microarray) scanners, sequencing machines, clinical notes or any other
biochemical,
biological, observational data, or the like. This step can be used to convert
the raw data into
a format that is better suited to analysis. In one example this is performed
in order to
normalize the raw data and thereby assist in ensuring the biomarker values
demonstrate
consistency even when measured using different techniques, different
equipment, or the like.
Thus, the goal of normalization is to remove the variation within the samples
that is not
directly attributable to the specific analysis under consideration. For
example, to remove
variances caused by differences in sample processing at different sites.
Classic examples of
normalization include z-score transformation for generic data, or popular
domain specific
normalizations, such as RMA normalization for microarrays.
[0383] However, it will also be appreciated that in some applications, such as
a single sample
experiment run on a single data acquisition machine, this step may not
strictly be necessary,
in which case the function can be a Null function producing an output
identical to the input.
[0384] In one example, the preferred approach is a paired function approach
over log
normalized data. Log normalization is a standard data transformation on
microarray data,
because the data follow a log-normal distribution when coming off the machine.
Applying a
log transform turns the data into process-friendly normal data.
[0385] At step 505 different groups are defined using the reference data.
[0386] Prior to this occurring, the processing system 201 optionally removes a
validation
subgroup of individuals from the reference data to allow the processing system
201 to
determine the candidate biomarkers using the reference data without the
validation subgroup

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 106 -
so that the validation subgroup can be subsequently used to validate the
candidate biomarkers
or signatures including a number of the candidate biomarkers. Thus, data from
the validation
subgroup is used to validate the efficacy of the candidate or signature
biomarkers in
identifying the presence, absence, degree, stage, severity, prognosis or
progression of any one
or more of the conditions to ensure the potential or signature biomarkers are
effective.
[0387] In one example, this is achieved by having the processing system 201
flag individuals
within the validation subgroup or alternatively store these in either an
alternative location
within the database 211 or an alternative database to the reference data. The
validation
subgroup of individuals is typically selected randomly and may optionally be
selected to
include individuals having different phenotypic traits. When a validation
subgroup of
individuals is removed, the remaining individuals will simply be referred to
as reference data
for ease throughout the remaining description.
[0388] The reference data (i.e., excluding the validation subgroup) are
classified into groups.
The groups may be defined in any appropriate manner and may be defined based
on any one
or more of an indication of a presence, absence, degree, stage, severity,
prognosis or
progression of a condition, other tests or assays, or measured biomarkers
associated with the
individuals.
[0389] For example, a first selection of groups may be to identify one or more
groups of
individuals suffering from SIRS, one or more groups of individuals suffering
ipSIRS, one or
more groups of individuals suffering inSIRS, and one or more groups of healthy
individuals.
Further groups may also be defined for individuals suffering from other
conditions. The
groups may include overlapping groups, so for example it may be desirable to
define groups
of healthy individuals and individuals having SIRS, with further being defined
to distinguish
inSIRS patients from ipSIRS patients, as well as different degree of inSIRS or
ipSIRS, with
these groups having SIRS in common, but each group of patients differing in
whether a
clinician has determined the presence of an infection or not. Additionally,
further
subdivision may be performed based on phenotypic traits, so groups could be
defined based
on gender, ethnicity or the like so that a plurality of groups of individuals
suffering from a
condition are defined, with each group relating to a different phenotypic
trait.
[0390] It will also be appreciated, however, that identification of different
groups can be
performed in other manners, for example on the basis of particular activities
of biomarkers

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 107 -
within the biological samples of the reference individuals, and accordingly,
reference to
conditions is not intended to be limiting and other information may be used as
required.
[0391] The manner in which classification into groups is performed may vary
depending on
the preferred implementation. In one example, this can be performed
automatically by the
processing system 201, for example, using unsupervised methods such as
Principal
Components Analysis (PCA), or supervised methods such as k-means or Self
Organizing
Map (SOM). Alternatively, this may be performed manually by an operator by
allowing the
operator to review reference data presented on a Graphical User Interface
(GUI), and define
respective groups using appropriate input commands.
[0392] At step 510 biomarkers are filtered based on their ability to
distinguish between the
groups. This process typically examines the activity of the reference
biomarkers for
individuals within and across the groups, to identify reference biomarkers
whose activities
differ between and hence can distinguish groups. A range of different analysis
techniques can
be utilized including, for example, regression or correlation analysis
techniques. Examples of
the techniques used can include established methods for parametized model
building such as
Partial Least Squares, Random Forest or Support Vector Machines, usually
coupled to a
feature reduction technique for the selection of the specific subset of the
biomarkers to be
used in a signature.
[0393] Such techniques are known and described in a number of publications.
For example,
the use of Partial Least Squares is described in "Partial least squares: a
versatile tool for the
analysis of high-dimensional genomic data" by Boulesteix, Anne-Laure and
Strimmer,
Korbinian, from Briefings in Bioinformatics 2007 vol 8. no. 1, pg 32-44.
Support Vector
machines are described in "LIBSVM: a library for support vector machines" by
Chang, C.C.
and Lin, C.J. from ACM Transactions on Intelligent Systems and Technology
(TIST), 2011
vol 2, no. 3,pg 27. Standard Random Forest in R language is described in
"Classification and
Regression by random Forest" by Liaw, A. and Wiener, M., in R news 2002, vol2,
no. 3, pg
18-22.
[0394] The analysis techniques are implemented by the processing system 201,
using
applications software, which allows the processing system 201 to perform
multiple ones of
the analysis techniques in sequence. This is advantageous as the different
analysis techniques
typically have different biases and can therefore be used to identify
different potential

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 108 -
biomarkers that can distinguish the groups, thereby reducing the risk of
clinically relevant
biomarkers being overlooked.
[0395] In one example, the process involves filtering out any biomarkers that
demonstrate a
correlation with the groups, and hence with the condition, that is below a
certain correlation
threshold, such as 0.3.
[0396] At step 515 derived biomarkers are generated from the filtered
reference biomarkers
using one or more functions. The nature of the derived biomarkers and the
functions used
will vary depending upon the preferred implementation. For example, functions
can include
division, subtraction, multiplication, addition of two markers, sigmoidal
functions applied to
the product of two biomarkers, negative logs of the division of two
biomarkers, least-squares
regression applied to two vectors of markers to produce a function (equation)
output,
concordance correlation coefficient of two vectors of categorical biomarkers,
or the like.
[0397] In general, the function is selected based on a number of rules. These
rules can
include: utility, functions that provide the best results; interpretability,
functions that can be
understood in terms of biological function; output, functions that produce
informative
outputs; simplicity; performance assessment; least number of biomarkers for
best
performance; number of biomarkers at a statistical overfitting threshold or
the like.
[0398] In one example, the preferred function is division, with the resulting
biomarkers being
different ratios. It will be appreciated that the division can be performed in
multiple different
ways, so that for three biomarkers, nine different derived biomarkers can be
determined.
[0399] At step 520 a performance measure is determined for each of the
candidate
biomarkers, including the filtered reference biomarkers and any derived
markers. The
performance measure may be of any suitable form and typically includes a
correlation or
performance explained measure that is indicative of a correlation of the
corresponding
biomarker and its ability to distinguish between groups. In one example, the
performance
function used to determine the performance measure is a standard univariate
statistical test
over all candidate biomarkers. Other examples however include a t-test, a non-
parametric
equivalent or area under receiver operator curve, chi squared or regression
analyses or their
equivalents, extensions or derivatives may be used.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 109 -
[0400] The outcome of the applying the performance function to each in the
candidate
selection step is a ranked list of biomarkers, with the top N ranked
biomarkers proceeding to
the next stage. The biomarkers falling below this threshold are no longer
considered. The
threshold applied may be an absolute number or proportion of all biomarkers,
or determined
by performance, such as a p value < 0.05. The threshold should be chosen to
contain a
sufficiently large number of biomarkers to bias towards including sufficiently
independent
biomarkers (i.e., low mutual correlation).
[0401] At step 525, the processing system 201 selects a next two candidate
biomarkers based
on the performance measure and on a mutual correlation. In this regard, two
markers that are
highly correlated with each other in terms of the context of the condition
will not necessarily
improve the ability to distinguish a particular presence, absence, degree or
prognosis of the
condition any more than a single one of the markers. Accordingly, it is
typical to select
biomarkers that have a high performance measure in respect of the condition,
but which have
a mutual correlation that falls below a mutual correlation threshold. The
mutual correlation
threshold used will vary depending upon the preferred implementation, and is
typically
selected to be as low as possible, as described above. Examples of the manner
in which the
biomarkers are selected will be described in more detail below with respect to
Figures 6A
and 6B.
[0402] At step 530, the processing system 201 determines a performance of a
next candidate
biomarker combination. In this regard, the processing system 201 will use a
combining
function such as addition, to combine the biomarker values of the selected
candidate
biomarkers and use this to determine and indicator value based on the
combination of
biomarker values. The performance can be determined in any suitable manner,
such as using
statistical measurements, correlation measurements, concordance measurements
or aggregate
performance measurements such as averages. In one particular example, the
performance
measure is a 'variance explained' (VE). A VE of "1" means that using the
biomarkers, you
can perfectly classify/predict the disease. A VE of "0.8" means that your
markers account for
80% of the result in practice.
[0403] Accordingly, at step 535 the processing system 201 compares the
performance of the
indicator to a performance threshold and determines if this is exceeded at
step 540. In the
event that the threshold is exceeded, this indicates that the selected
combination of markers

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 1 1 0 -
provides the required degree of discrimination allowing the presence, absence,
degree or
prognosis of the condition to be determined.
[0404] In the event that the threshold is not exceeded at step 540, it is
determined if all
combinations have been considered at step 545. In this regard, it is possible
that multiple
different combinations of the two selected biomarkers to be tried, so if each
possible
combination has not been considered, the processing system 201 returns to step
530 to
determine the performance of a next candidate biomarker combination. In this
regard, the
combinations used will typically be ordered in terms of preference, so that
preferred
combinations are tried first, with less preferred combinations being tried
only in the event
that preferred combinations prove unsuccessful.
[0405] Once all candidate biomarker combinations have been considered for the
two
candidate biomarkers selected, and if the performance threshold has still not
been exceeded,
the process moves onto step 550 to compare the current number of candidate
biomarkers
being considered to a limit. In this regard, the limit is used to control the
overall number of
biomarkers in the biomarker signature, thereby minimizing signature size and
hence the cost
of performing the associated measurements and diagnosis.
[0406] If the limit has not been exceeded an additional biomarker is added
based on the
correlation and performance at step 560, with the process moving onto step 530
to determine
a performance of the next candidate biomarker combination. Otherwise if a
limit has been
reached, then an alternative next two candidate biomarkers are selected at
step 525. Thus,
this process allows additional candidate biomarkers to be progressively
included, with a
combination of the multiple candidate biomarkers being compared to the
performance
threshold, to determine if the required performance is met. If this is not
achieved before the
number of candidate biomarkers reaches the limit, the process is recommenced
using two
different candidate biomarkers.
[0407] Once the performance threshold has been exceeded at step 540, the
selected candidate
biomarkers can be defined as signature biomarkers for inclusion in a biomarker
signature for
the one or more conditions at step 565.
[0408] It should be noted that before the biomarker signature is finalized at
step 565,
additional checks might be performed, to ensure that the candidate biomarkers
included in the
signature should not be excluded for any reason. For example, candidate
biomarkers might

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 1 1 1 -
be excluded for cost considerations as some combinations of candidate
biomarkers may cost
more than others. For example, a larger number of biomarkers may cost more
than a smaller
number, and the additional cost may not be justified by a small improvement in
performance.
Alternatively, the cost might be increased if multiple different tests are
required in order to
measure required biomarker values.
[0409] Biomarkers might also be excluded from use for legal reasons, for
example if their
use is restricted for approval or intellectual property reasons. Some
biomarkers may be
difficult to measure from a technical perspective, for example very low
expression in vivo,
which increases variability and therefore reduces robustness.
[0410] The performance of each biomarker combination panel may also include
some
variability, typically expressed as confidence intervals around the reported
performance.
Although a point estimate for one panel may be higher than for another, if the
difference
given the variability is not significant, the combinations may be considered
equivalent.
[0411] Once a particular combination of signature biomarkers has been defined,
at step 570
the processing system 201 can determine an indicator value range associated
with each
group. In particular, the range of reference biomarker values for the
signature biomarkers
within each group are used to calculate indicator value ranges for each group.
These can then
be compared to an indicator value calculated for a biological subject having
an unknown
presence absence, degree or progression of the at least one condition and used
to identify a
group to which the subject would belong and hence the presence, absence,
degree or
progression of the condition.
[0412] Thus, the above-described process iteratively assesses the biomarkers,
initially
selecting two biomarkers, with various combinations of these being considered
to determine
if these have the required performance for use in diagnosing the presence,
absence, degree or
progression of a condition. In the event that the required performance is not
provided,
additional biomarkers can be added and further combinations tried. Thus the
process can
consider three biomarkers, four biomarkers, five biomarkers, six biomarkers,
seven
biomarkers, eight biomarkers, nine biomarkers or more. Typically this is
performed to a
limit which may be defined based for example on the number of biomarkers that
can
practically be measured within given cost or process parameters. In the event
that the

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 112 -
required performance is not obtained the process moves onto to select
alternative candidate
biomarkers with this being repeated.
[0413] Thus it will be appreciated that the above process initially selects
those biomarkers
which have the suitable performance and which are not highly correlated on the
basis that
these provide the maximum performance. The ability of these biomarkers to
distinguish is
then tested and in the event that this is insufficient, further biomarkers can
be added to a
limit. If this still does not provide the required discriminatory performance
alternative
biomarkers can be selected.
[0414] The process of selecting two candidate biomarkers at step 525 can be
achieved in any
number of ways depending upon the preferred implementation and examples of
this will now
be described with reference to Figures 6A and 6B.
[0415] In the example of Figure 6A, at step 600 biomarkers are grouped
according to their
mutual similarity. Thus, highly correlated biomarkers are put together in
common groups.
Biomarkers within the group are ranked at step 610 based on their performance
measure in
terms of their correlation with the condition, with the highest ranked
biomarkers from two of
the groups being selected at step 620 to define the next two candidate
biomarkers. It will be
appreciated if additional candidate biomarkers are required, these can be
selected from
different groups to the first two candidate biomarkers.
[0416] An alternative process is shown in Figure 6B. In this example, at step
650 biomarkers
are ranked based on their performance at discriminating the condition(s). At
step 660 a next
highest biomarker is selected, with the remaining biomarkers being re-ranked
on the
combination of their similarity with the selected biomarker, for example using
mutual
information, as well as their performance. The next highest biomarker is then
selected at step
680 with this process being repeated as required.
[0417] Accordingly, it will be appreciated that the above-described processes
provide
mechanisms for selecting a combination of biomarkers, and more typically
derived
biomarkers, that can be used to form a biomarker signature, which in turn can
be used in
diagnosing the presence, absence or degree of at least one condition or in
providing a
prognosis of at least one condition. In this regard, the biomarker signature
defines the
biomarkers that should be measured (i.e., the signature biomarkers), how
derived biomarker
values should be determined for measured biomarker values, and then how
biomarker values

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 113 -
should be subsequently combined to generate an indicator value. The biomarker
signature
can also specify defined indicator value ranges that indicate a particular
presence, absence,
degree or prognosis of one or more conditions.
[0418] An example of the method of using a biomarker signature described above
will now
be described with reference to Figure 7.
[0419] In this example, at step 700 a plurality of measured biomarker values
are measured
for a biological subject whose condition is unknown, with these typically
being provided to
the processing system 201, for example by download from measuring equipment or
the like.
[0420] After any required processing, such as normalization or the like, at
step 710, the
processing system 201 applies one or more functions to the measured biomarker
values to
determine any required derived biomarker values. A derived biomarker value and
another
biomarker value (i.e., another derived biomarker value or a measured biomarker
value) are
combined to generate an indicator value, which can then be displayed or
otherwise used in
determining the presence, absence, degree or prognosis of one or more
conditions. Thus, this
can involve simply displaying the indicator value, allowing an assessment to
be made by a
medical practitioner or alternatively may involve further processing, such as
comparing the
indicator to defined indicator value ranges that indicate a particular
presence, absence, degree
or prognosis of one or more conditions, with the results of the comparison
being displayed.
[0421] Accordingly, in the above-described method the biomarker signature
defines the
biomarker values that need to be measured and/or derived, allowing the
processing system
201 to automatically generate an indicator value based on received measured
biomarker
values. Once this has been completed, the processing system 201 can compare
the indicator
value to the indicator value ranges, and either display results of the
comparison, or alternative
interpret the results of the comparison, allowing an indicator to be displayed
that is indicative
of the presence, absence, degree or prognosis of a condition. This can then be
used by a
medical practitioner as required in performing a medical diagnosis of the
biological subject.
[0422] Using the above-described methods it has been identified that the use
of ratios of
"immune system biomarkers" is particularly beneficial when assessing a
likelihood of a
biological subject having a presence, absence, degree or prognosis of at least
one medical
condition.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-114-
104231 As used herein, the term "immune system biomarker" refers to a
biomarker of the
host's immune system that is altered, or whose level of expression is altered,
as part of an
inflammatory response to damage or pathogenic insult, including metabolic,
toxic,
neurotoxic, iatrogenic, thermal or chemical insults, illustrative examples of
which include
trauma, surgery, drugs including chemotherapeutic drugs, radiation, disease
including
pathogenic infection, metabolic disease and ischemia, as well as foreign or
implanted
substances.
[0424] The term "immune system", as used herein, refers to cells, molecular
components and
mechanisms, including antigen-specific and non-specific categories of the
adaptive and
innate immune systems, respectively, that provide a defense against damage and
insults and
matter, the latter comprised of antigenic molecules, including but not limited
to tumors,
pathogens, and self-reactive cells.
[0425] The term "innate immune system" refers to a host's non-specific
reaction to insult to
include antigen-nonspecific defense cells, molecular components and mechanisms
that come
into action immediately or within several hours after exposure to almost any
insult or antigen.
Elements of the innate immunity include for example phagocytic cells
(monocytes,
macrophages, dendritic cells, polymorphonuclear leukocytes such as
neutrophils,
reticuloendothelial cells such as Kilpffer cells, and microglia), cells that
release inflammatory
mediators (basophils, mast cells and eosinophils), natural killer cells (NK
cells) and physical
barriers and molecules such as keratin,
mucous, secretions, complement proteins,
immunoglobulin M (IgM), acute phase proteins, fibrinogen and molecules of the
clotting
cascade, and cytokines. Effector compounds of the innate immune system include
chemicals
such as lysozymes, IgM, mucous and chemoattractants (e.g., cytokines or
histamine),
complement and clotting proteins.
[0426] The term "adaptive immune system" refers to antigen-specific cells,
molecular
components and mechanisms that emerge over several days, and react with and
remove a
specific antigen. The adaptive immune system develops throughout a host's
lifetime. The
adaptive immune system is based on leukocytes, and is divided into two major
sections: the
humoral immune system, which acts mainly via immunoglobulins produced by B
cells, and
the cell-mediated immune system, which functions mainly via T cells.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-115-
104271 Accordingly, in one example, an indicator is determined that correlates
to a ratio of
immune system biomarkers, which can be used in assessing a likelihood of a
biological
subject having a presence, absence, degree or prognosis of at least one
medical condition.
[0428] In this example, the method includes determining a pair of biomarker
values, each
biomarker value being a value measured or derived for at least one
corresponding immune
system biomarker of the biological subject and being at least partially
indicative of a
concentration of the immune system biomarker in a sample taken from the
subject.
[0429] The biomarker values are used to determine a derived biomarker value
using the pair
of biomarker values, the derived biomarker value being indicative of a ratio
of concentrations
of the pair of immune system biomarkers.
[0430] Thus, if the biomarker values are the concentrations of the biomarkers,
then the
derived biomarker value will be based on a ratio of the biomarker values.
However, if the
biomarker values are related to the concentrations of the biomarkers, for
example if they are
logarithmically related by virtue of the biomarker values being based on PCR
cycle times, or
the like, then the biomarker values may be combined in some other manner, such
as by
subtracting the cycle times to determine a derived biomarker value indicative
of a ratio of the
concentrations.
[0431] The derived biomarker is then used to determine the indicator, either
by using the
derived biomarker value as an indicator value, or by performing additional
processing, such
as comparing the derived biomarker value to a reference or the like, as will
be described in
more detail below.
[0432] In any event, combining biomarker values to determine a ratio of
concentrations of
immune system biomarkers, and then using this to determine an indicator allows
indicators to
be determined for use in determining a likelihood of a subject suffering from
a range of
different conditions, depending on the immune system biomarkers selected,
which as it will
be appreciated can be performed using the above described process.
[0433] A number of further features will now be described.
[0434] In one example, the process involves determining a first derived
biomarker value
using a first pair of biomarker values, the first derived biomarker value
being indicative of a
ratio of concentrations of first and second immune system biomarkers,
determining a second

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 116 -
derived biomarker value using a second pair of biomarker values, the second
derived
biomarker value being indicative of a ratio of concentrations of third and
fourth immune
system biomarkers and determining the indicator by combining the first and
second derived
biomarker values. Thus, in this example, two pairs of derived biomarker values
can be used,
which can assist in increasing the ability of the indicator to reliably
determine the likelihood
of a subject having a condition.
[0435] The derived biomarker values could be combined using a combining
function such as
an additive model; a linear model; a support vector machine; a neural network
model; a
random forest model; a regression model; a genetic algorithm; an annealing
algorithm; a
weighted sum; a nearest neighbor model; and a probabilistic model.
[0436] In one example, the indicator is compared to an indicator reference,
with a likelihood
being determined in accordance with results of the comparison. The indicator
reference is
typically derived from indicators determined for a number of individuals in a
reference
population. The reference population typically includes individuals having
different
characteristics, such as a plurality of individuals of different sexes; and/or
ethnicities, with
different groups being defined based on different characteristics, with the
subject's indicator
being compared to indicator references derived from individuals with similar
characteristics.
The reference population can also include a plurality of healthy individuals,
a plurality of
individuals suffering from at least one diagnosed medical condition, a
plurality of individuals
showing clinical signs of at least one medical condition and/or first and
second groups of
individuals, each group of individuals suffering from a respective diagnosed
medical
condition.
[0437] It will be appreciated that the individuals selected will depend on the
intended use of
the indicator. In particular, when the indicator is for use in determining the
likelihood that a
biological subject has a specific medical condition, the sample population
includes
individuals presenting with clinical signs of the specific medical condition,
individuals
diagnosed with the specific medical condition and healthy individuals. This
ensures that the
assessment of indicator validity applies regardless of not or whether the
individual has the
specific condition or not.
[0438] It will also be appreciated that the sample population could also
include a plurality of
individuals of different sexes, ethnicities, ages, or the like, allowing the
control value ranges

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 117 -
to be common across populations. However, this is not essential, and
alternatively control
value thresholds could be established that are specific to a particular sub-
set of the
population. In this case, it would be necessary to ensure that the control
value threshold
ranges used are appropriate for the subject under consideration.
[0439] The indicator can also be used for determining a likelihood of the
subject having a
first or second condition, in other words to distinguish between the
conditions. In this case,
this would typically be achieved by comparing the indicator to first and
second indicator
references, the first and second indicator references being indicative of
first and second
conditions and determining the likelihood in accordance with the results of
the comparison.
In particular, this can include determining first and second indicator
probabilities using the
results of the comparisons and combining the first and second indicator
probabilities, for
example using a Bayes method, to determine a condition probability
corresponding to the
likelihood of the subject having one of the conditions. In this situation the
first and second
conditions could include two medical conditions, or a single medical condition
and a healthy
condition.
[0440] In this case, the first and second indicator references are
distributions of indicators
determined for first and second groups of a reference population, the first
and second group
consisting of individuals diagnosed with the first or second condition
respectively. In this
regard, this can be achieved by determining first and second groups of
individuals, each
group of individuals having a presence or absence of a diagnosed medical
condition and
determining first and second indicator references for the first and second
groups respectively.
This allows the indicator to be used to distinguish between first and second
conditions, which
could include different medical conditions, as well as healthy conditions. It
will also be
appreciated that whilst two groups are described, this is not essential and
three or more
groups could also be defined.
[0441] The process is usually performed using at least one electronic
processing device, such
as a suitably programmed computer system or the like.
[0442] In this case, the electronic processing device typically obtains at
least two pairs of
measured biomarker values, either by receiving these from a measuring or other
quantifying
device, or by retrieving these from a database or the like. The processing
device then
determines a first derived biomarker value indicative of a ratio of
concentrations of first and

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 118 -
second immune system biomarkers and a second derived biomarker value
indicative of a
ratio of third and fourth immune system biomarkers. The processing device then
determines
the indicator by combining the first and second derived biomarker values.
[0443] The processing device can then generate a representation of the
indicator, for example
by generating an alphanumeric indication of the indicator, a graphical
indication of a
comparison of the indicator to one or more indicator references or an
alphanumeric indication
of a likelihood of the subject having at least one medical condition.
[0444] The method would also typically include obtaining a sample taken from
the biological
subject, the sample including polynucleotide expression products and
quantifying at least
some of the polynucleotide expression products within the sample to determine
the pair of
biomarker values. This can be achieved using any suitable technique, and will
depend on the
nature of the immune system biomarkers.
[0445] For example, if the indicator is based on a ratio of concentrations of
the
polynucleotide expression products, this process would typically include
quantifying
polynucleotide expression products by amplifying at least some polynucleotide
expression
products in the sample, determining an amplification amount representing a
degree of
amplification required to obtain a defined level of each of a pair of
polynucleotide expression
products and determining the indicator by determining a difference between the
amplification
amounts. In this regard, the amplification amount is generally a cycle time, a
number of
cycles, a cycle threshold and an amplification time.
[0446] In this case, the method includes determining a first derived biomarker
value by
determining a difference between the amplification amounts of a first pair of
polynucleotide
expression products, determining a second derived biomarker value by
determining a
difference between the amplification amounts of a second pair of
polynucleotide expression
products and determining the indicator by adding the first and second derived
biomarker
values.
[0447] As previously discussed, the at least two immune system biomarkers have
a mutual
correlation in respect of the at least one condition that lies within a mutual
correlation range,
the mutual correlation range being between 0.9 and the indicator has a
performance value
greater than or equal to a performance threshold representing the ability of
the indicator to

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 119 -
diagnose the presence, absence, degree or prognosis of the at least one
condition, the
performance threshold being indicative of an explained variance of at least
0.3.
[0448] Typically the mutual correlation range is one of 0.8; 0.7; 0.6;
0.5; 0.4; 0.3;
0.2; and, 0.1.
[0449] Typically each immune system biomarker has a condition correlation with
the
presence, absence, degree or prognosis of the at least one condition that lies
outside a
condition correlation range, the condition correlation range being between
0.3 and more
typically 0.9; 0.8; 0.7; 0.6; 0.5; and, 0.4. Typically the performance
threshold is
indicative of an explained variance of at least one of 0.4; 0.5; 0.6; 0.7;
0.8; and, 0.9.
[0450] The above-described method has been used to identify 1650 biomarkers of

inflammatory response syndromes (also referred to interchangeably herein as
"IRS
biomarkers" or "IRS immune system biomarkers"), which are useful for assisting
in
distinguishing: (1) between SIRS affected subjects (i.e., subjects having
inSIRS or ipSIRS)
and healthy subjects or subjects not affected by SIRS; (2) between subjects
with inSIRS and
subjects with ipSIRS; and/or (3) between subjects with different stages of
ipSIRS (e.g.,
sepsis, severe sepsis and septic shock). Based on this identification, the
present inventors
have developed various methods, compositions, apparatus and kits, which take
advantage of
these biomarkers to provide an indicator for use in diagnosing the presence,
absence or
degree of at least one condition, or for prognosing at least one condition,
wherein the at least
one condition is selected from a healthy condition (e.g., a normal condition
or one in which
inSIRS and inSIRS are absent), inSIRS, ipSIRS, or a stage of ipSIRS (e.g., a
stage of ipSIRS
with a particular severity, illustrative examples of which include mild
sepsis, severe sepsis
and septic shock). In advantageous embodiments, the methods and kits involve
monitoring
the expression of IRS biomarker genes in cells of the immune systems,
including blood cells
(e.g., immune cells such as leukocytes), which may be reflected in changing
patterns of RNA
levels or protein production that correlate for example with the presence of
active disease or
response to disease.
[0451] The IRS biomarkers are expression products of genes (also referred to
interchangeably herein as "IRS biomarker genes" or IRS immune system biomarker
genes"),
including polynucleotide and polypeptide expression products. As used herein,
polynucleotide expression products of IRS biomarker genes are referred to
herein as "IRS

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 120 -
biomarker polynucleotides." Polypeptide expression products of the IRS
biomarker genes are
referred to herein as "IRS biomarker polypeptides." The term "gene", as used
herein, refers
to a stretch of nucleic acid that codes for a polypeptide or for an RNA chain
that has a
function. While it is the exon region of a gene that is transcribed to form
RNA (e.g., mRNA),
the term "gene" also includes regulatory regions such as promoters and
enhancers that govern
expression of the exon region.
[0452] Suitably, the IRS biomarker genes are selected from the group
consisting of: PRKCZ,
SKI, RER1, TAS1R1, VAMP3, AGTRAP, VPS13D, KLHDC7A, NBL1//Clorf151, MDS2,
RCAN3, LDLRAP1, MAN1C1, SH3BGRL3, DHDDS, HCRTR1, CCDC28B, LCK,
ZNF362, THRAP3, PPIE//CCDC25, CAP], CTPS, Clorf84, FAAH, DMBX1, CYP4B1,
BTF3L4, LRRC42, Clorf175//TTC4, TMEM61, FPGT//TNNI3K, ACADM, SPATA1,
EPHX4, RPAP2, RPL5//SNORA66//SNORD21//FAM69A, RTCD1, SLC30A7,
RNPC3//AMY2B, CELSR2, AHCYL1, CEPT1//DR14M2, CHIA, LIX1L, UPF0627,
MRPS21, TNFAIP8L2, SMCP, DCST1, RAG1AP1, Clorf182, HAPLN2, NTRK1, CD1E,
TOW4OL//NR1I3, POU2F1, TIPRL, SFT2D2, CACNA1E, SMG7, OCLM, RGS2,
ZC3H11A//RP11-74E24.2, MFSD4, IL20, RPS6KC1, Clorf95, ARE], GALNT2,
TNFRSF4, NADK, FLI14100//Clorf86, GPR153, RERE, SLC2A7, SDHB, RNF186,
DDOST, GPN2, RPA2, PEF1, PTP4A2, TRIM62, PHC2, LSM10, MRPS15, RRAGC,
COL9A2, TESK2, NRD1, KTI12, CC2D1B, YIPF1, JAK1, SLC35D1, DIRAS3, ZZZ3,
GNG5, ZNHIT6, ODF2L, SEP15, BARHL2, GCLM, CLCC1//GPSM2//Clorf62, SORT],
SLC16A4, PHTF1, RSBN1, DENND2C//BCAS2, CD58, SPAG17//WDR3, REG4//NBPF7,
RP11-
9412.2//NBPF16//NBPF11//NBPF15//NBPF8//NBPF20//NBPF10//NBPF14//NBPF1//LOC10
0288142//NBPF12//KIAA1245//LOC100290137, APH1A, POGZ, TDRKH, THEM4,
S100A11, CRNN, SPRR2C, S100Al2, S100A8, GATAD2B//PLIN2, DENND4B, PBXIP1,
PYG02, SHC1, DCST2, GBA//GBAP, ASH1L, PIT], MEF2D, AIM2, COPA, DEDD,
TADA1L, GPA33, CD247, F5, PIGC, KIAA0040, TOR1AIP2//TOR1AIP1//IFRG15,
STX6//KI4A1614, EDEM3, UCHL5, DENND1B, DDX59, KIF21B, ARL8A, CYB5R1,
MYBPH, CHI3L1, PIK3C2B//LOC100130573, NUAK2, NUCKS1, FAIM3, PLXNA2,
SLC30A1, LPGAT1, ANGEL2, RAB3GAP2//AURKAPS1//AURKA//SNORA36B, TP53BP2,
NVL, TMEM63A, PARP1, ITPKB, TARBP1, CHML, AKT3, SMYD3, AHCTF1, OR1C1,
NCOA1, HADHB, ABHD1//PREB, SPAST, 5LC30A6//DDX50, CRIPT, MSH2, FOXN2,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 121 -
CCDC104, VRK2, AHSA2//USP34, OTX1, AFTPH, CEP68, PLEK, ANXA4, MXD1,
NAGK, SMYD5//NOTO, MTHFD2, TTC31, SEA/L614F, TMSB10, SH2D6, GNLY,
KCNIP3, CN1VM4, CNNM3, ZAP 70, LIPT1//MRPL30, MAP4K4, IL1R2, IL1R1,
IL18R1, POLR1B, CHCHD5, IL1RN, PSD4, DDX18, INSIG2,
TMEA1177//LOC100125918, RALB, PROC, GPR17//LOC100291428//LIMS2, IMP4,
FAM123C, ACVR2A, MBD5, LYPD6B, SLC4A10, UBR3, HAT], ITGA6, ZAK,
OSBPL6, PLEKHA3, ZC3H15, COL3A1, GLS, OBFC2A, COQ10B, MARS2, CFLAR,
N0P58, FAM117B, CYP20A1, FASTKD2, PIKFYVE, C2orf62, SLC11A1, AGFG1,
CHRNG, EIF4E2, TRPM8, LRRFIP1, GAL3ST2, TMEA118, LAPTM4A, SF3B14,
TP53I3, UNQ2999, GPR113//SELI, MPV17, PPM1G, NLRC4, CDC42EP3, HNRPLL,
COX7A2L, KCNG3, CALA12//C2orf61, BCL11A, XP01, NAT8B, DUSP11, MOGS,
SNRNP200, SEA/L614C, MITD1, ILIA, SLC35F5, CCDC93, CLASP], SAP130, YSK4,
GTDC1, ORC4L, NR4A2//FLI46875, DPP4, GALNT3, SCN7A, FRZB, STK17B,
CLK1//PPIL3, MPP4, IN080D, KLF7, FAM119A, NGEF, ARL4C, RAB17, HDLBP,
LRRN1, SETD5, IRAK2, C3orf42, TSEN2, NR2C2//MRPS25, UBE2E1, C3orf35, SNRK,
ZNF197, GNAI2, ALAS], PRKCD, CACNA1D, PXK, PTPRG, ATXN7, 5LC35A5,
SLC15A2, CCDC48, DNAJC13, CLDN18, GYG1, SELT, MED12L, RAP2B, MYNN,
ABCF3, VPS8, HRG, EIF4A2//SNORA4, LPP, CCDC50, LOC152217, TADA3L,
SEC13, TIMP4, METTL6, DAZL//DAZ4//DAZ3//DAZ2, SATB1//TBC1D5, SCN10A,
SEC22C, ZDHHC3, ZDHHC3, SLC6A20, UQCRC1, PRICAR2A, IMPDH2, CCDC71,
UBA7, CAMKV, WDR82, LAIOD3, FOXP1, MORC1, ATG3, GSK3B//LOC100129275,
HCLS1, KPNA1, PTPLB, C3orf22, RPN1, KIAA1257//ACAD9//LOC100132731, FOXL2,
MECOM, PLD1, GNB4, MRPL47, KLHL6, THPO, ETV5, BCL6//LOC100131635,
ATP13A5, TMEA144, KI4A1530, TACC3, CNO, BST], KLF3, TMEA133//DCAF4L1,
KIT, ENAM, FAM47E//STBD1, ENOPH1, PDLIM5, CCDC109B//HIGD1A//CCDC13,
EGF, PCDH10, RAB33B, TMEA1184C, RBM46, GRIA2, C4orf39, KLHL2, TLL1, F11,
SLBP, HAUS3//POLN, PPARGC1A, TLR10, C4orf34, TXK, RPL21P44, KDR, RCHY1,
CNOT6L, PLAC8, HPSE, GPRIN3, PPA2, COL25A1, C4orf3, QRFPR, MFSD8,
MAP9, PDGFC, TKTL2, ACSL1, SUB1//TMEA1183A, CARD6, MCCC2, TNP01,
PDE8B, PAPD4, THBS4, FAM151B, RASGRF2, SNX2, LMNB1//PCIF1, MEGF10,
LEAP2, TCF7, KDM3B, CXXC5, SLC4A9, ANKHD1-EIF4EBP3//ANKHD1//EIF4EBP3,
KIAA0141, GRPEL2, MFAP3, GABRA6, GABRA1, DOCK2, RANBP171/USP12,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 122 -
ERGIC1, ATP6V0E1//SNORA74B, ZNF346, NSD1, CLP1M1L, UGT3A1, GDNF,
TTC33, hCG 2039148, MOCS2, SLC38A9, CCDC125, ANKRA2, HAPLN1, CCNH,
TMEM161B, MBLAC2, MCTP1, TICAM2//TMED7//TMED7-TICAM2, KIF3A, C5orf15,
SKP1, CXCL14, KLHL3, CD14, YIPF5, LAPS, DCTN4, CCDC69, ATOX1, TIMD4,
ADAM19, SLIT3, RNF44, DOK3, MGAT4B//SQS1M1, C5orf45//SQS1M1, RASGEF1C,
MGAT1, IRF4, HIVEP1, E2F3, HIST1H4I, HIST1H2BM, MOG, ZNRD1//NCRNA00171,
TRIM15, HCG27, BAT2//SNORA38, CYP21A2, ITPR3, MAPK14, MAPK13, PNPLA1,
SFRS3, CDKN1A, FOXP4, CUL9, RUNX2, ZNF451, SOBP, C6orf182, KIAA1919,
RWDD1, KPNA5, TPD52L1, ARG1, RAB32, ARID1B, SLC22A3, SERPINB1, C6orf146,
GCM2, ATX1V1, DCDC2//KAAG1, HIST1H3I, HIST1H4L, GABBR1, RNA243, DDAH2,
CLIC], NEU], RXRB, VPS52, TCP11, CLPS, PGC, ZNF318, YIPF3, MRPL14,
PLA2G7, PK_HD1, IL17F, HTR1B, GABRR2, UBE2I1, BACH2, MCM9, VNN1,
IL2ORA, FLI27255, T, RPS6KA2, HGC6.3, UNC84A//C7orf20, SDK], ZDHHC4,
C7orf26, GLCCI1//tcag7.903, GPNMB, CCDC126, WIPF3//ZNRF2//L0C441208,
GPR141, STARD3NL, POU6F2, CDC2L5, ZMIZ2, UPP1, ZNF273, KCTD7//RABGEF1,
RABGEF1//tcag7.967//tcag7.951//KCTD7//L0C100293333,
CCDC132,
PVRIG//PILRB//STAG3, PILRB//PVRIG//STAG3, C7orf51, GNB2, LRRC17, LRRN3,
CFTR, LSM8, LUC7L2, MGAM//L0C100124692, GIMAP7, INSIG1, RBM33, ICA1,
FAM126A, HIBADH, TRIL, SCRN1, ELM01, INHBA, CAMK2B, NPC1L1,
DDC//LOC100129427, NSUN5//NSUN5B//NSUN5C, CLDN3, C7orf23//DMTF1, SRI,
BET], MCM7, GA TS, ATXN7L1//RINT1//EFCAB10, KIAA1549, SLC37A3, SMARCD3,
MLL3//BAGE2, CLN8, MSRA, PIWIL2, NEFM//L0C100129717, EPHX2, LEPROTL1,
MAK16//C8orf41, AP3M2, FNTA, SGK196, UBE2V2, FLI46365, SNTG1, TRIM55,
C8orf45, PREX2, PLEK_HF2, BAALC//FLI10489, TTC35, MTBP, ZHX2, RNF139, TG,
DENND3//C8orf60, INFRSF10D, TRIM35, GSR, WHSC1L1, PCMTD1//PXDNL,
NCOA2, TRAM1//L0C286190, RUNX1T1, EXT1, DDEF1IT1, CDC37L1, UBE2R2,
UBAP1//KIF24, GALT, RGP1//GBA2, TGFBR1, C9orf6//IKBKAP, IMAGE5303689,
ATP6V1G1, TLR4, SET, MRPL41, C9orf68, HAUS6//SCARNA8, KLHL9, C9orf82,
NDUFB6//DFFB, SIT], FAM108B1, TRPM6, FRMD3, 5LC28A3, BICD2, C9orf84,
AKNA, MEGF9, C5, GOLGA1//SCAL SH2D3C, FAM102A, FLI10232, ASB6, BAT2L,
EDF], FBXW5, ClOorf18, FBX018, GATA3, CUGBP2, VIM, STAM, WAC, BAMBI,
ZNF4871/L0C439911, ALOX5, WDFY4, SRGN, CCDC109A, FAM149B1//FAM149B2,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 123 -
MINPP 1, PTEN//PTENP1, ENTPD1//C 1 Oorfl 31, ABCC2, SFXN2, SHOC2, ACSL5,
BCCIP//DHX32, FAM188A, CUBN, SVIL//hCG 1783494, FAM13C//PHYHIPL, ATAD1,
ANKRD22, FLI34077, COX15, ERLIN1, ACTR1A, ABLIM1, RAB11FIP 2, C 1 Oorf84,
PRDX3, C 1 Oorf119, NSMCE4A, TALD01//INTS8, TNNT3, FXC 1 , PDE3B, DNAJC24,
PTPRJ//0R4B1, C 1 1 orf31, TMEM109, CD6, CD5, TMEM1 38, POLR2G, TMEM-179B,
NAT]], OTUB1, RBM14//RBM4, AIP, PPFIA1, IL18BP//NUMA1, C 1 1 orf30, C 1 1
orf82,
TMEM-126B, C 1 1 orf7 3, PIWIL4, L0C1001 32 686, PAFAH1B2, UBE4A, TRAPPC4,
SC5DL, VWA5A//0R10D1P, STT3A, VP S26B, TRIM21, ZBED5, SAAL1, FANCF,
LIN7C, PHF21A, CUGBP 1, OSBP, CYBASC3, TUT], SLC25A45, LTBP3, EIF1AD,
GAB2, CREBZF, PICALM, SLC36A4, CCDC82, KIAA1826, MPZL3, MPZL2, H2AFX,
SIAE, ZBTB44, HSN2, ADIPOR2, NCAPD2//SCARNA10//FADS1, PTPN6, CLEC4D,
CDKN1B, GOLT1B, FAR2, FGD4, TMEM-106C, TMBIM6, C 1 2orf62, PRR1 3//PCBP 2,
DGKA, COQ10A, TSPAN31, CDK4/MARCH9/C3HC4, LEMD3, IRAK3, TMTC3,
AC TR6, TC TAT] , PXMP2//PGAM5, DCP1B, SLC2A3//SLC2A14, C3AR1, PLBD1,
TM7SF3, ASB8//PHB, LMBR1L, FMNL3//PRPF4OB , AAAS, NFE2, GPR84, CD63,
SARNP//DNAJC14, NACA, CDK4//TSPAN31, TMBIM4//L0C1001 33322, IL22, LIN7A,
HAL, APPL2, GLTP, GIT2, VPS29, PP TC7, DDX54//CCDC42B, SLC24A6, SDS,
RBM19, MED13L, C 1 2orf49, FBX021, WSB2, TAOK3, CIT, RAB35, RPLPO, PXN,
TRIAP 1 , SFRS9, POPS, UNQ188 7, C 1 2orf43, ANAPC5 , KDM2B, MORN3,
TMEA1120B//RHOF, L0C338 799, DIABLO//B3GNT4, VPS33A, CLIP 1 , PITPNM2,
EIF2B 1, CCDC92, NCOR2, DHX3 7, DDX51, POLE, GOLGA 3, ZMYM2,
SPATA13//C1QTNF9, NUPL1, PAN3//EEF1A1//CHCHD2, ALOX5AP, EEF1DP3, KL,
UFM1, NARG1L, ITM2B, FNDC3A, CDADC1 , ARL11, LM07, DNAJC3, TM9SF2,
CLYBL, PCCA, ABHD1 3, LAMP 1 , TMC03, UPF3A, ZMYM5//ZMYM2,
ZDHHC2 0//LOC728 099, PARP4, MTMR6//L0C646482, HSPH1, N4BP2L2//CG030,
ELF], LCP 1, KPNA3, C 1 3orfl , DLEU2//DLEU2L, GUCY1B2, INTS6, DACH1,
TBC1D4, EDNRB, UGGT2, GPR183, LIG4, ANKRD10, RASA3, RNASE2//L0C643332,
RPGRIP 1, IRF9, TSSK4, C 1 4orf21, SCFD1, FANCM, ABHD12B, PTGDR,
FBX034//KIAA0831, C 1 4orf101, ACTR10, ARID4A, JKAMP, HIF1A, SYNE2, EXD2,
5LC39A9, SFRS5, PCNX, SIPAlL 1//SNORD56B//LOC1454 741/L0C28 3567, YLPM1,
BATF, FLVCR2//RP524, GPR65, TDP 1, EVL, ZNF839, TDRD9, INF2, PLD4,
MTA1//LOC64 73] 0//LOC100128343 , NDRG2, DAD]//0R611, SLC7A8, IP04, TM9SF1,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 124 -
ADCY4, RIPK3, EAPP, BAZ1A, NFKBIA, SEC23A, Cl4orf104, Cl4orf138, SOS2, NIN,
PYGL, CNIH, DHRS7, WDR89, ACTN1, NUMB, Cl4orf43, ABCD4, KIAA0317, NEK9,
ANGEL], SPTLC2, SERPINA6, DICER], BCL11B, ANKRD9, PPP1R13B, AKT1,
BRF1 , TUBGCP 5 , SNRPN//SNURF//IP W//SNORD 1 1 6-1 6//SNORD 1 1 6-18//SNORD 1
1 6-
2 1//SNORD1 1 6-22//SNORD 1 1 6-1 7//SNORD 1 1 6-19//PAR5//PAR-SN//SNORD 1 1 6-

2//SNORD1 1 6-25//SNORD 1 1 6-2 6//SNORD 1 0 7//SNORD 1 15-1 21/SNORD 1 1 5-
5//SNORD 1 15-
6//SNORD1 1 5-9//SNORD 1 1 6-1 1//SNORD 1 1 6-1 2//SNORD 1 1 6-1 3//SNORD 1 1
6-
28//SNORD1 1 6-4//SNORD64//PAR1//SNORD 1 09A//SNORD 1 09B//SNORD 1 1 6-
6//SNORD1 1 6-3//SNORD 1 1 6-9//SNORD 1 1 5-1 3//SNORD1 1 5-1//SNORD 1 1 5-
14//SNORD1 1 5-1 5//SNORD 1 15-2 1//SNORD 1 15-1 0//SNORD 1 15-7//SNORD 1 15-
I 6//SNORD1 1 5-40//SNORD 1 1 5-42//SNORD 1 15-1 1//SNORD 1 15-29//SNORD 1 15-
34//SNORD1 1 5-3 6//SNORD 1 1 5-4//SNORD 1 1 5-43//HBI1-52-24//SNORD 1 1 6-
5//SNORD 1 1 6-
7//SNORD1 15-2 6//SNORD 1 15-3 0//SNORD 1 1 6-1 5//SNORD 1 1 6-8//SNORD 1 1 5-
2//SNORD1 15-39//SNORD 1 1 6-1 4//SNORD 1 1 6-2 0//SNORD 1 1 5-8//SNORD 1 1 5-
3//SNORD1 15-38//SNORD 1 15-4 1//SNORD 1 15-22//SNORD 1 1 5-44//SNORD 1 1 6-
1//SNORD1 15-1 7//SNORD 1 15-18//SNORD 1 15-19//SNORD 1 15-2 0//SNORD 1 1 6@,
APBA 2 ,
MTMR15//MTMR10, RYR3, BAHD1, CHP, JMID7-PLA2G4B//JMID7//PLA2G4B,
HAUS2, Cl5orf63//SERF2, B2M, TRIM69, PLDN, SQRDL, GALK2, USP8, GLDN,
MAPK6, LACTB, RAB8B, APH1B, USP3//LOC100130855, SNX1,
LBXCOR1//PIAS1//CALAIL4, NE01, MPI, FBX022//FBX0220S, RCN2, FAH, IL16,
ABHD2, SLCO3A1, MCTP2, MEF2A//LYSMD4,
NIPA2//CYFIP1,
HERC2//HERC2P2//HERC2P3//L0C440248, MTMR10//MTMR15, Cl5orf24, SLC12A6,
LPCAT4, IN080, 01P5, ZFP106, CDAN1, SPG11//ISLR, SPPL2A,
GNB5//LOC100129973, MY05A, ARPP19, RAB27A, CCPG1//PIGB//DYX1C1, BNIP2,
CA12, FAM96A, KIAA0101//CSNK1G1, TLE3, PARP6, NPTN, MAN2C1, IMP3,
MTHFS, 5T20//C15orf37, TMC3, AP3B2, Cl5orf40, WDR73, NTRK3, DET1, TM2D3,
WDR90, RHOT2//FBXL16, TMEAI204, CRAMP1L//HN1L, MAPK8IP3, TBL3, TSC2,
KCTD5//PR00461//PDPK1, CLUAP1, DNASE1, DNAJA3, CP110, Cl6orf62, LYRAll,
METTL9, EEF2K, POLR3E, PLK1, PRKCB, IL21R//L0C283888, SULT1A2//SULT1A1,
ATXN2L,
LAT//SPNS1//NPIPL2//LOC728 741//L0C 7 3 0 15 3//NPIPL3//SPIN1//LOC
728888//LOC1 0 028
9169//LOC728734//L0C729602//LOC100288442//LOC100288332, K1F22,
MAZ,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 125 -
CORO1A//L0C606724, ITGAL, SRCAP//SNORA30, ZNF646//ZNF668, Cl6orf67,
TMEA1188, LPCAT2, CETP, CKLF, CMTM1//CKLF, TMEA1208, CTCF, THAP11,
NUTF2, EDC4, SLC7A6//SLC7A60S, PRA1T7, SNTB2, VPS4A, DDX19B//DDX19A,
CHST4, HP//HPR, PLCG2, KLHL36, KIAA0182, BANP//RUNDC2C, TRAPPC2L,
SPG7, CDK10, TCF25, AFG3L1, LUC7L, AXIN1, JAVD8, LAIF1, UNKL, UNKL,
CLCN7, MRPS34, RNPS1, NLRC3, TRAP1//DNASE1, ADCY9, COR07, C16orf72,
RRN3//L0C653390//L0C730092//LOC 100131998,
XYLT1//LYRA12//ZC3H11A,
DCUN1D3//LYR711 , IGSF6//METTL9,
CDR2//RRN3//LOC 100131998//LOC653390,
COG 7, GGA2, NSMCE1, GTF3C1, CCDC101//L0C388242, C16orf54, KCTD13,
SEPT], ZNF764//ZNF747, Cl6orf58//LOC100128371, ITFG1, ABCC11//LONP2,
NUDT21, BBS2//0GFOD1, CSNK2A2, GOT2, FAM96B, FHOD1//SLC9A5,
ATP6V0D1//L0C100132855, GFOD2, SLC12A4, DPEP3, DPEP2, CHTF8//HAS3,
COG8//PDF, TERF2, AARS, ST3GAL2, VAC14//L0C100130894, AP1G1, WDR59,
CTRB2//CTRB1, TAF1C//ADAD2, FBX031, ZCCHC14, FAM38A, CENPBD1, TIMM22,
RPA1, DPH1//0VCA2, SGSM2, ARRB2, L0C100130950, DNAH2, PIGL, TRPV2,
MPRIP, DRG2, ALKBH5//FL113773, SMCR7, WSB1, TAOK1, CPD, SUZ12P, RNF135,
ZNF830, TAF15, GGNBP2, LASP1, PSMD3, CDC6, NBR2, TMUB2,
MGC57346//C17orf69, NSF//LOC728806, GOSR2, NPEPPS//TBC1D3F//L0C440434,
KPNB1, CDK5RAP3, ATP5G1, UBE2Z, XYLT2//L0C100130580, NOG, DGKE, AKAP1,
TMEA149//CLTC//MIR21, CLTC, CA4, C17orf64, DCAF7, PITPNC1,
NOL11//SNORA38B, MAP2K6, COG], CD300A, TMEA1104, MRPS7, KI4A0195,
TSEN54, LLGL2, L0C100134934//CDK3, MFSD11, SEPT9, TNRC6C, TMC8,
ENGASE, RPTOR, GPS1, FN3KRP, TBCD, GEMIN4, GLOD4, 5LC43A2, PRPF8,
5MG6//C17orf6, METT10D//LOC284009, SHPK, TAX1BP3, P2RX5, MYBBP1A//SPNS2,
PELP1, PFN1, ZNF232, DHX33, DERL2, NLRP1//LOC728392, ASGR2,
NEURL4//GPS2//D45234E, ZBTB4, TP53, VAMP2, PIK3R5, ELAC2,
NCOR1//C20orf191//LOC 100131704, ZNF287,
TOM1L2//L0C246315,
GRAP//SNORD3B-1//SNORD3B-2//L0C400581, ALDOC, SDF2, RAB34, PHF12,
NUFIP2, OMG, EVI2B, Cl7orf66//RSL24D1, SYNRG//LOC100131822, PLXDC1,
CACNB1, PGAP3, MED24, NR1D1//THRA, CCR7, STAT5B//STAT5A, FAM134C,
VAT], DUSP3, C17orf65//ASB16, UBTF, GPATCH8, MAP3K14//L0C100133991,
OSBPL7, SLC35B1, TOB1, COX11//TOM1L1, VEZFl, SFRS1//FLI44342, SEPT4,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 126 -
MED13//L0C100129112, LIMD2//MAP3K3, STRADA, FTS,13, CD79B, ICAM2, ERN],
TEX2,
LRRC37A 3//LRRC 37A2//LRRC37A//ARL 17P 1//LRRC37A4//LOC 1 0 0294335//LOC644397
,
GNA13, WIPI1//ARSG, FAM20A, NAT9, GGA3, H3F3B//H3F3C, EXOC7, SFRS2,
TMC6//L0C100131096, USP36, CD7, RAB31, VAPA, SEH1L, HQ0644/PR00644,
RNMT, RNF138, GALNT1, ELP2, PIK3C3, SLC14A2, ME2, SERPINB2//SERPINB10,
ZNF407, ZNF236, NFATC1//LOC100127994, ENOSF1//TYMS, MYOM1, AFG3L2,
ABHD3, OSBPL1A, CDH2, DSC1, PSTPIP2, C18orf32, MBD2//SNORA37, PIGN,
TMX3, PQLC1, GZW, ARID3A, CIRBP, DAZAP1, SPPL2B, NFIC, VA Vi,
ARHGEF18//LOC100128573, STXBP2//L0C554363//LOC100131801, Cl9orf59, ZNF317,
ILF3, SMARCA4, PRKCSH, IER2, CCDC130, DCAF15, IL27RA, KLF2, SIN3B,
DDA1, GTPBP3, FAM129C, FCH01, ARRDC2, IFI30, Cl9orf60, CRTC1//MAML2,
RFXANK//MEF2B//LOC729991, ZNF101,
ZNF738,
ZNF257//ZNF492//ZNF99//ZNF98//L0C646864, C19orf2, KIAA0355//FL121369, USF2,
TMEM-147, LIN37//PSENE1V, C19orf55, TBCB//POLR2I, ZNF382, ZNF568, ZNF420,
ZNF383, CCDC97, ZNF574, CD177, ZNF230//ZNF222, VASP, GRWD1, FLT3LG,
ZNF175, NCRNA00085, PPP2R1A, ZNF808//ZNF578//ZNF611, LENG8, FCAR, RPL28,
U2AF2, L0C100288114//MGC9913, ZFP28, ZNF460, ZNF549, ZNF211,
ZNF5871/ZNF417, ZNF274, ZNF544, ZNF8, TRIM28, C19orf6, C19orf34, GNG7, AES,
EEF2//SNORD37, PLIN5//LRG1, PLIN3, PTPRS, SAFB2//SAFB, RANBP3,
GTF2F1//L0C100130856, XAB2, ELAVL1, ADAMTS10, FBXL12, DNMT1, TYK2,
KEAP1, KRI1, TMEM205//hCG 29977, ZNF563, MAN2B1//MORG1, C19orf56, DHPS,
TNP02//SNORD41, LPHN1, NDUFB7, AKAP8, AKAP8L, CHERP//C19orf44//CALR3,
INSL3//JAK3, IL12RB1, UPK1A, TYROBP, ZNF529, ZNF461, ZNF607, YIF1B, PRR13,
CEACAM4, PLAUR, TRAPPC6A, ERCC1//CD3EAP, RTN2, SYMPK, PGLYRP1,
NO SIP, PNKP, NKG7, FPR1, ZNF28, OSCAR, MBOAT7, LILRA5, LILRA4,
ZNF550//ZNF549, ZNF416, ZNF256, ZNF329, FAM110A, ITPA, CDC25B, CDS2,
CRLS1, CSRP2BP, SEC23B, 5LC24A3, HCK, ASXL1, ACSS2, C20orf4, TGIF2,
C20orf24//SLA2, RPN2//EEF1A2, CTNNBL1, ACTR5, PPP1R16B, DHX35, PLCG1,
MYBL2, 5Y51//5Y51-DBNDD2//DBNDD2, DNTTIP1, CTSA, WP9//LOC100128028,
DDX27, SLC9A8, RNF114, PTPN1, TSHZ2, PFDN4, CSTF1, CASS4, GNAS,
C20orf177, CDH26, C20orf197, L0C284757, ARFGAP1, PRPF6, NSFL1C, SIRPD,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 127 -
SIRPG//SIRPA, R]'/F24, RASSF2, TMX4, JAG], C20orf74, C20orf3, C20orf112,
CDK5RAP1, AHCY, GGT7, EDEM2, RBM39//L0C643167, BLCAP, SERINC3//TTPAL,
ZNF335, ELM02, B4GALT5, DPM1, ZFP64, ZNF217, CTSZ, SYCP2, PSMA7, DID01,
YTHDF1, CHODL, BACH1, C2lorf41//BACH1, ILlORB, IFNAR1, IFNGR2, SON,
MORC3//DOPEY2, DYRK1A, KCNI15, ETS2, RRP1B, PFKL, TRPM2, ADARB1,
SAMSN1//L0C388813, N6AMT1, SYNA TMEM50B, KCNE1, PRDM15, C2CD2,
WDR4, U2AF1, CSTB, UBE2G2//SUM03, PTTGlIP, POFUT2, MCM3AP,
IL17RA//CECR7, C22orf37, LZTR1, PPIL2//YPEL1, CYTSA, SNRPD3//C22orf13, NF2,
LIMK2, SLC5A1, MCM5, NCF4, GGA1, SH3BP1//PDXP, POLR2F//LOC100131530,
APOBEC3A//APOBEC3B, APOBEC3D, ATF4, CACNA1I, ZC3H7B, CCDC134, TSPO,
NUP50, TBC1D22A//LOC100289878, RP3-402G11.5, SAPS2, NCAPH2, BID, SLC25A1,
KLHL22//KRT18, PI4KA//PI4KAP1//PI4KAP2//LOC100293141, MAPK1, ZNF70, TPST2,
SF3A1//CCDC157, PES1, PIK3IP1, PATZ1, C22orf30, IL2RB, CSNK1E//L0C400927,
UNC84B, CBX7//LOC100128400, RPS19BP1, MKL1//KI4A1659, RANGAP1, TCF20,
LDOC1L, UNQ6126, TUBGCP6, SBF1//SBF1P1, MSL3, MOSPD2, BMX//HNRPDL,
PDHAl, YY2, PDK3, GK//GK3P//FTL//L0C652904, CXorf59, ATP6AP2,
USP9X//USP9Y, RP2, USP11, RBM3, FTSJ1, WAS, PLP2, TSPYL2//GPR173,
MAGED2, UBQLN2, NLGN3, ACRC, UPRT, CXorf26, ATP7A, DIAPH2,
CSTF2//RAD21, AMCX3, AMCX5, GPRASP1, TMEM31, TBC1D8B, MID2,
DOCK]], LONRF3, UBE2A, SH2D1A, OCRL, SLC25A14, HPRT1, CD4OLG, AFF2,
55R4//IDH3G, FAM50A, DKC11/SNORA36A//SNORA56, ARSD, KALI, CTPS2,
RPS6KA3, BCOR, MA0B//NAT13, ZNF41, OTUD5, KCND1, ZMYM3, MAGT1,
BRWD3, TRI1T2B, GLA, MORF4L2, PSMD10, ACSL4, LAMP2, CUL4B, ODZ1,
ELF4, RAP2C, FAM127B//FAM127C//FAM127A, TMEM185A, ARD1A, IRAK],
DNASE1L1//RPL10, SH3KBP1, Mitochondrial, Mitochondrial, CCNL2, INPP5B, TLR5,
ADRB3//GOT 1L 1 , NOC2L//SAMD11//LOC401010 and SHFM1 (hereafter referred to
interchangeably herein as "the full list of IRS immune system biomarker genes"
or "full list
IRS biomarker genes").
[0453] The methods, compositions, apparatus and kits of the present invention
take
advantage of the IRS biomarkers broadly described above and elsewhere herein
to provide an
indicator for use in diagnosing the presence, absence or degree of the at
least one condition
selected from a healthy condition (e.g., a normal condition or one in which
inSIRS and

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 128 -
inSIRS are absent), inSIRS, ipSIRS or a stage of ipSIRS (e.g., a stage of
ipSIRS with a
particular severity such as mild sepsis, severe sepsis and septic shock), or
in providing a
prognosis of the at least one condition, which may involve: (a) determining a
plurality of IRS
biomarker values, each IRS biomarker value being indicative of a value
measured or derived
for at least one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) IRS biomarker
of a biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values (also referred to herein as a "biomarker signature"), the indicator
being at least
partially indicative of the presence, absence, degree or prognosis of the at
least one condition,
wherein: (i) at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) IRS
biomarkers have a mutual
correlation in respect of the at least one condition that lies within a mutual
correlation range,
the mutual correlation range being between 0.9; and (ii) the indicator has a
performance
value greater than or equal to a performance threshold representing the
ability of the indicator
to diagnose the presence, absence or degree of the at least one condition, or
to provide a
prognosis for the at least one condition, the performance threshold being
indicative of an
explained variance of at least 0.3.
[0454] In advantageous embodiments, the diagnostic or prognostic methods,
compositions,
apparatus and kits of the present invention involve: (1) determining a
plurality of measured
IRS biomarker values, each measured IRS biomarker value being a measured value
of an IRS
biomarker of the biological subject; and (2) applying a function to at least
one of the
measured IRS biomarker values to determine at least one derived IRS biomarker
value, the at
least one derived IRS biomarker value being indicative of a value of a
corresponding derived
IRS biomarker. The function suitably includes at least one of: (a) multiplying
two IRS
biomarker values; (b) dividing two IRS biomarker values; (c) adding two IRS
biomarker
values; (d) subtracting two IRS biomarker values; (e) a weighted sum of at
least two IRS
biomarker values; (f) a log sum of at least two IRS biomarker values; and (g)
a sigmoidal
function of at least two IRS biomarker values.
[0455] In some embodiments, the diagnostic or prognostic methods,
compositions, apparatus
and kits involve: determining at least one derived IRS biomarker value
corresponding to a
ratio of two measured IRS biomarker values. In these examples, the diagnostic
or prognostic
methods, apparatus and kits suitably include combining at least two IRS
biomarker values to
determine an indicator value representing the indicator and in illustrative
examples of this

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 129 -
type, the at least two IRS biomarker values are combined using a combining
function (e.g.,
any one or more of: an additive model; a linear model; a support vector
machine; a neural
network model; a random forest model; a regression model; a genetic algorithm;
an annealing
algorithm; weighted sum; a nearest neighbor model; and a probabilistic model).
Suitably, the
diagnostic or prognostic methods, apparatus and kits include: (a) determining
a first derived
IRS biomarker value, the first derived IRS biomarker value being a ratio of
first and second
measured IRS biomarker values; (b) determining a second derived IRS biomarker
value, the
second derived IRS biomarker value being a ratio of third and fourth measured
IRS
biomarker values; and (c) adding the first and second derived IRS biomarker
values to
generate an indicator value.
[0456] In some embodiments, the methods, compositions, kits and apparatus of
the present
invention are useful for diagnosing that inSIRS or a healthy condition is
present or absent in
the biological subject, which suitably involve: (a) determining a plurality of
IRS biomarker
values, each IRS biomarker value being indicative of a value measured or
derived for at least
one IRS biomarker of a biological subject; (b) determining the indicator using
a combination
of the plurality of IRS biomarker values, the at least one indicator being at
least partially
indicative of the presence, absence, degree or prognosis of the at least one
condition selected
from inSIRS and a healthy condition, wherein: (i) at least two IRS biomarkers
have a mutual
correlation in respect of the at least one condition that lies within a mutual
correlation range,
the mutual correlation range being between 0.9; and (ii) the indicator has a
performance
value greater than or equal to a performance threshold representing the
ability of the indicator
to diagnose the presence, absence or degree of the at least one condition, or
to provide a
prognosis for the at least one condition, the performance threshold being
indicative of an
explained variance of at least 0.3, wherein at least one of the at least two
IRS biomarkers is
selected from a first IRS biomarker group and wherein at least one other of
the at least two
IRS biomarkers is selected from a second IRS biomarker group, wherein the
first IRS
biomarker group consists of polynucleotide and/or polypeptide expression
products from
group A IRS biomarker genes as defined herein, and wherein the second IRS
biomarker
group consists of polynucleotide and/or polypeptide expression products from
group B IRS
biomarker genes as defined herein.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 130 -
[0457] In other embodiments, the methods, apparatus and kits are useful for
diagnosing that
ipSIRS or a healthy condition is present or absent in the biological subject,
which suitably
involve: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value being
indicative of a value measured or derived for at least one IRS biomarker of a
biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from ipSIRS and a
healthy
condition, wherein: (i) at least two IRS biomarkers have a mutual correlation
in respect of the
at least one condition that lies within a mutual correlation range, the mutual
correlation range
being between 0.9; and (ii) the indicator has a performance value greater
than or equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group C IRS
biomarker genes,
as defined herein, and wherein the second IRS biomarker group consists of
polynucleotide
and/or polypeptide expression products from group D IRS biomarker genes as
defined herein.
[0458] In still other embodiments, the methods, apparatus and kits are useful
for diagnosing
that inSIRS or ipSIRS is present or absent in the biological subject, which
suitably involve:
(a) determining a plurality of IRS biomarker values, each IRS biomarker value
being
indicative of a value measured or derived for at least one IRS biomarker of a
biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from inSIRS and
ipSIRS, wherein:
(i) at least two IRS biomarkers have a mutual correlation in respect of the at
least one
condition that lies within a mutual correlation range, the mutual correlation
range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 131 -
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group E IRS
biomarker genes as
defined herein, and wherein the second IRS biomarker group consists of
polynucleotide
and/or polypeptide expression products from group F IRS biomarker genes as
defined herein.
[0459] In other embodiments, the methods, apparatus and kits are useful for
diagnosing that
inSIRS or ipSIRS is present or absent in the biological subject, which
suitably involve: (a)
determining a plurality of IRS biomarker values, each IRS biomarker value
being indicative
of a value measured or derived for at least one IRS biomarker of a biological
subject; (b)
determining the indicator using a combination of the plurality of IRS
biomarker values, the at
least one indicator being at least partially indicative of the presence,
absence, degree or
prognosis of the at least one condition selected from inSIRS and ipSIRS,
wherein: (i) at least
four IRS biomarkers have a mutual correlation in respect of the at least one
condition that lies
within a mutual correlation range, the mutual correlation range being between
0.9; and (ii)
the indicator has a performance value greater than or equal to a performance
threshold
representing the ability of the indicator to diagnose the presence, absence or
degree of the at
least one condition, or to provide a prognosis for the at least one condition,
the performance
threshold being indicative of an explained variance of at least 0.3, wherein
at least one of the
at least four IRS biomarkers is selected from a first IRS biomarker group,
wherein at least
one other of the at least four IRS biomarkers is selected from a second IRS
biomarker group,
wherein at least one other of the at least four IRS biomarkers is selected
from a third IRS
biomarker group, and wherein at least one other of the at least four IRS
biomarkers is
selected from a fourth IRS biomarker group, wherein the first IRS biomarker
group consists
of polynucleotide and/or polypeptide expression products from group G IRS
biomarker genes
as defined herein, wherein the second IRS biomarker group consists of
polynucleotide and/or
polypeptide expression products from group H IRS biomarker genes as defined
herein,
wherein the third IRS biomarker group consists of polynucleotide and/or
polypeptide
expression products from group I IRS biomarker genes as defined herein, and
wherein the
fourth IRS biomarker group consists of polynucleotide and/or polypeptide
expression
products from group J IRS biomarker genes as defined herein.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 132 -
[0460] In still other embodiments, the methods, apparatus and kits are useful
for diagnosing
that mild sepsis or severe sepsis is present or absent in the biological
subject, which suitably
involve: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value being
indicative of a value measured or derived for at least one IRS biomarker of a
biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from mild sepsis
and severe sepsis,
wherein: (i) at least two IRS biomarkers have a mutual correlation in respect
of the at least
one condition that lies within a mutual correlation range, the mutual
correlation range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group K IRS
biomarker genes as
defined herein, and wherein the second IRS biomarker group consists of
polynucleotide
and/or polypeptide expression products from group L IRS biomarker genes as
defined herein.
[0461] In still other embodiments, the methods, apparatus and kits are useful
for diagnosing
that mild sepsis or septic shock is present or absent in the biological
subject, which suitably
involve: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value being
indicative of a value measured or derived for at least one IRS biomarker of a
biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from mild sepsis
and septic shock,
wherein: (i) at least two IRS biomarkers have a mutual correlation in respect
of the at least
one condition that lies within a mutual correlation range, the mutual
correlation range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 133 -
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group M IRS
biomarker genes
as defined herein, and wherein the second IRS biomarker group consists of
polynucleotide
and/or polypeptide expression products from group N IRS biomarker genes as
defined herein.
[0462] In still other embodiments, the methods, apparatus and kits are useful
for diagnosing
that severe sepsis or septic shock is present or absent in the biological
subject, which suitably
involve: (a) determining a plurality of IRS biomarker values, each IRS
biomarker value being
indicative of a value measured or derived for at least one IRS biomarker of a
biological
subject; (b) determining the indicator using a combination of the plurality of
IRS biomarker
values, the at least one indicator being at least partially indicative of the
presence, absence,
degree or prognosis of the at least one condition selected from severe sepsis
and septic shock,
wherein: (i) at least two IRS biomarkers have a mutual correlation in respect
of the at least
one condition that lies within a mutual correlation range, the mutual
correlation range being
between 0.9; and (ii) the indicator has a performance value greater than or
equal to a
performance threshold representing the ability of the indicator to diagnose
the presence,
absence or degree of the at least one condition, or to provide a prognosis for
the at least one
condition, the performance threshold being indicative of an explained variance
of at least 0.3,
wherein at least one of the at least two IRS biomarkers is selected from a
first IRS biomarker
group and wherein at least one other of the at least two IRS biomarkers is
selected from a
second IRS biomarker group, wherein the first IRS biomarker group consists of
polynucleotide and/or polypeptide expression products from group 0 IRS
biomarker genes as
defined herein, and wherein the second IRS biomarker group consists of
polynucleotide
and/or polypeptide expression products from group P IRS biomarker genes as
defined herein.
[0463] As used herein, the terms "diagnosis", "diagnosing" and the like are
used
interchangeable herein to encompass determining the likelihood that a subject
will develop a
condition, or the existence or nature of a condition in a subject. These terms
also encompass
determining the severity of disease or episode of disease, as well as in the
context of rational
therapy, in which the diagnosis guides therapy, including initial selection of
therapy,
modification of therapy (e.g., adjustment of dose or dosage regimen), and the
like. By

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 134 -
"likelihood" is meant a measure of whether a biological subject with
particular measured or
derived biomarker values actually has a condition (or not) based on a given
mathematical
model. An increased likelihood for example may be relative or absolute and may
be
expressed qualitatively or quantitatively. For instance, an increased
likelihood may be
determined simply by determining the subject's measured or derived biomarker
values for at
least two IRS biomarkers and placing the subject in an "increased likelihood"
category, based
upon previous population studies. The term "likelihood" is also used
interchangeably herein
with the term "probability".
[0464] In some embodiments, the biomarkers, including IRS biomarkers, are
obtained from a
biological sample. The term "biological sample" as used herein refers to a
sample that may
be extracted, untreated, treated, diluted or concentrated from an animal. The
biological
sample is suitably a biological fluid such as whole blood, serum, plasma,
saliva, urine, sweat,
ascitic fluid, peritoneal fluid, synovial fluid, amniotic fluid, cerebrospinal
fluid, tissue biopsy,
and the like. In certain embodiments, the biological sample contains blood,
especially
peripheral blood, or a fraction or extract thereof Typically, the biological
sample comprises
blood cells such as mature, immature or developing leukocytes, including
lymphocytes,
polymorphonuclear leukocytes, neutrophils, monocytes, reticulocytes,
basophils,
coelomocytes, hemocytes, eosinophils, megakaryocytes, macrophages, dendritic
cells natural
killer cells, or fraction of such cells (e.g., a nucleic acid or protein
fraction). In specific
embodiments, the biological sample comprises leukocytes including peripheral
blood
mononuclear cells (PBMC). By "obtained from" is meant to come into possession.
Biological
or reference samples so obtained include, for example, nucleic acid extracts
or polypeptide
extracts isolated or derived from a particular source. For instance, the
extract may be isolated
directly from a biological fluid or tissue of a subject.
[0465] The term "nucleic acid" or "polynucleotide" as used herein includes
RNA, mRNA,
miRNA, cRNA, cDNA mtDNA, or DNA. The term typically refers to a polymeric form
of
nucleotides of at least 10 bases in length, either ribonucleotides or
deoxynucleotides or a
modified form of either type of nucleotide. The term includes single and
double stranded
forms of DNA or RNA.
[0466] "Protein", "polypeptide" and "peptide" are used interchangeably herein
to refer to a
polymer of amino acid residues and to variants and synthetic analogues of the
same.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 135 -
[0467] In some embodiments, biomarker signatures are determined through
analysis of
measured or derived IRS biomarker values for IRS biomarkers of one or more
control
subjects that have or do not have a condition. These biomarkers are referred
to herein as
"reference IRS biomarkers". In specific examples, individual control subjects
are selected
from "healthy control subjects", "non-healthy control subjects", "SIRS control
subjects",
"inSIRS control subjects", "ipSIRS control subjects", "control subjects with a
particular stage
of ipSIRS", illustrative examples of which include "mild sepsis control
subjects", "severe
sepsis control subjects" and "septic shock control subjects", etc.), which are
also referred to
herein as control groups (e.g., "healthy control group", "non-healthy control
group", "SIRS
control group", "inSIRS control group", "ipSIRS control group", "ipSIRS stage
group",
illustrative examples of which include "mild sepsis control group", "severe
sepsis control
group", and "septic shock control group", etc.).
[0468] Suitably, an individual measured or derived IRS biomarker value
corresponds to the
level or amount of a respective IRS biomarker or to a function that is applied
to that level or
amount. As used herein the terms "level" and "amount" are used interchangeably
herein to
refer to a quantitative amount (e.g., weight or moles), a semi-quantitative
amount, a relative
amount (e.g., weight % or mole % within class), a concentration, and the like.
Thus, these
terms encompass absolute or relative amounts or concentrations of IRS
biomarkers in a
sample.
[0469] In some embodiments, the presence, absence, degree or prognosis of at
least one
condition in a biological subject is established by determining a plurality of
IRS biomarker
values, wherein each IRS biomarker value is indicative of a value measured or
derived for at
least one IRS biomarker in a biological sample obtained from the biological
subject. These
biomarkers are referred to herein as "sample IRS biomarkers". In accordance
with the present
invention, a sample IRS biomarker corresponds to a reference IRS biomarker
(also referred to
herein as a "corresponding IRS biomarker"). By "corresponding IRS biomarker"
is meant an
IRS biomarker that is structurally and/or functionally similar to a reference
IRS biomarker.
Representative corresponding IRS biomarkers include expression products of
allelic variants
(same locus), homologues (different locus), and orthologues (different
organism) of reference
IRS biomarker genes. Nucleic acid variants of reference IRS biomarker genes
and encoded
IRS biomarker polynucleotide expression products can contain nucleotide
substitutions,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 136 -
deletions, inversions and/or insertions. Variation can occur in either or both
the coding and
non-coding regions. The variations can produce both conservative and non-
conservative
amino acid substitutions (as compared in the encoded product). For nucleotide
sequences,
conservative variants include those sequences that, because of the degeneracy
of the genetic
code, encode the amino acid sequence of a reference IRS polypeptide.
[0470] Generally, variants of a particular IRS biomarker gene or
polynucleotide will have at
least about 40%, 45%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59% 60%,
61%,
62%, 63%, 64%, 65%, 66%, 67%, 68%, 69% 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%,

78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%,
93%,
94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to that particular
nucleotide
sequence as determined by sequence alignment programs known in the art using
default
parameters. In some embodiments, the IRS biomarker gene or polynucleotide
displays at
least about 40%, 45%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59% 60%,
61%,
62%, 63%, 64%, 65%, 66%, 67%, 68%, 69% 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%,

78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%,
93%,
94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to a nucleotide
sequence
selected from any one of SEQ ID NO: 1-1650, as summarized in Table 1.
[0471] Corresponding IRS biomarkers also include amino acid sequences that
display
substantial sequence similarity or identity to the amino acid sequence of a
reference IRS
biomarker polypeptide. In general, an amino acid sequence that corresponds to
a reference
amino acid sequence will display at least about 50, 51, 52, 53, 54, 55, 56,
57, 58, 59, 60, 61,
62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80,
81, 82, 83, 84, 85, 86,
97, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% or even up to 100%
sequence similarity or
identity to a reference amino acid sequence selected from any one of SEQ ID
NO: 1651-
3284, as summarized in Table 2.
[0472] In some embodiments, calculations of sequence similarity or sequence
identity
between sequences are performed as follows:
[0473] To determine the percentage identity of two amino acid sequences, or of
two nucleic
acid sequences, the sequences are aligned for optimal comparison purposes
(e.g., gaps can be
introduced in one or both of a first and a second amino acid or nucleic acid
sequence for
optimal alignment and non-homologous sequences can be disregarded for
comparison

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 137 -
purposes). In some embodiments, the length of a reference sequence aligned for
comparison
purposes is at least 30%, usually at least 40%, more usually at least 50%,
60%, and even
more usually at least 70%, 80%, 90%, 100% of the length of the reference
sequence. The
amino acid residues or nucleotides at corresponding amino acid positions or
nucleotide
positions are then compared. When a position in the first sequence is occupied
by the same
amino acid residue or nucleotide at the corresponding position in the second
sequence, then
the molecules are identical at that position. For amino acid sequence
comparison, when a
position in the first sequence is occupied by the same or similar amino acid
residue (i.e.,
conservative substitution) at the corresponding position in the second
sequence, then the
molecules are similar at that position.
[0474] The percentage identity between the two sequences is a function of the
number of
identical amino acid residues shared by the sequences at individual positions,
taking into
account the number of gaps, and the length of each gap, which need to be
introduced for
optimal alignment of the two sequences. By contrast, the percentage similarity
between the
two sequences is a function of the number of identical and similar amino acid
residues shared
by the sequences at individual positions, taking into account the number of
gaps, and the
length of each gap, which need to be introduced for optimal alignment of the
two sequences.
[0475] The comparison of sequences and determination of percentage identity or
percentage
similarity between sequences can be accomplished using a mathematical
algorithm. In certain
embodiments, the percentage identity or similarity between amino acid
sequences is
determined using the Needleman and Wunsch, (1970, 1 Mol. Biol. 48: 444-453)
algorithm
which has been incorporated into the GAP program in the GCG software package
(available
at http://www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix,
and a gap
weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or
6. In specific
embodiments, the percent identity between nucleotide sequences is determined
using the
GAP program in the GCG software package (available at http://www.gcg.com),
using a
NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length
weight of 1,
2, 3, 4, 5, or 6. An non-limiting set of parameters (and the one that should
be used unless
otherwise specified) includes a Blossum 62 scoring matrix with a gap penalty
of 12, a gap
extend penalty of 4, and a frameshift gap penalty of 5.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 138 -
[0476] In some embodiments, the percentage identity or similarity between
amino acid or
nucleotide sequences can be determined using the algorithm of E. Meyers and W.
Miller
(1989, Cabios, 4: 11-17) which has been incorporated into the ALIGN program
(version 2.0),
using a PAM120 weight residue table, a gap length penalty of 12 and a gap
penalty of 4.
[0477] The nucleic acid and protein sequences described herein can be used as
a "query
sequence" to perform a search against public databases to, for example,
identify other family
members or related sequences. Such searches can be performed using the NBLAST
and
)(BLAST programs (version 2.0) of Altschul, et at., (1990, J Mot Biol., 215:
403-10).
BLAST nucleotide searches can be performed with the NBLAST program, score =
100,
wordlength = 12 to obtain nucleotide sequences homologous to 53010 nucleic
acid molecules
of the invention. BLAST protein searches can be performed with the )(BLAST
program,
score = 50, wordlength = 3 to obtain amino acid sequences homologous to YYYYY
protein
molecules of the invention. To obtain gapped alignments for comparison
purposes, Gapped
BLAST can be utilized as described in Altschul et at., (1997, Nucleic Acids
Res, 25: 3389-
3402). When utilizing BLAST and Gapped BLAST programs, the default parameters
of the
respective programs (e.g., )(BLAST and NBLAST) can be used.
[0478] Corresponding IRS biomarker polynucleotides also include nucleic acid
sequences
that hybridize to reference IRS biomarker polynucleotides, or to their
complements, under
stringency conditions described below. As used herein, the term "hybridizes
under low
stringency, medium stringency, high stringency, or very high stringency
conditions"
describes conditions for hybridization and washing. "Hybridization" is used
herein to denote
the pairing of complementary nucleotide sequences to produce a DNA-DNA hybrid
or a
DNA-RNA hybrid. Complementary base sequences are those sequences that are
related by
the base-pairing rules. In DNA, A pairs with T and C pairs with G. In RNA, U
pairs with A
and C pairs with G. In this regard, the terms "match" and "mismatch" as used
herein refer to
the hybridization potential of paired nucleotides in complementary nucleic
acid strands.
Matched nucleotides hybridize efficiently, such as the classical A-T and G-C
base pair
mentioned above. Mismatches are other combinations of nucleotides that do not
hybridize
efficiently.
[0479] Guidance for performing hybridization reactions can be found in Ausubel
et at.,
(1998, supra), Sections 6.3.1-6.3.6. Aqueous and non-aqueous methods are
described in that

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 139 -
reference and either can be used. Reference herein to low stringency
conditions include and
encompass from at least about 1% v/v to at least about 15% v/v formamide and
from at least
about 1 M to at least about 2 M salt for hybridization at 42 C, and at least
about 1 M to at
least about 2 M salt for washing at 42 C. Low stringency conditions also may
include 1%
Bovine Serum Albumin (BSA), 1 mM EDTA, 0.5 M NaHPO4 (pH 7.2), 7% SDS for
hybridization at 65 C, and (i) 2 x SSC, 0.1% SDS; or (ii) 0.5% BSA, 1 mM
EDTA, 40 mM
NaHPO4 (pH 7.2), 5% SDS for washing at room temperature. One embodiment of low

stringency conditions includes hybridization in 6 x sodium chloride/sodium
citrate (SSC) at
about 45 C, followed by two washes in 0.2 x SSC, 0.1% SDS at least at 50 C
(the
temperature of the washes can be increased to 55 C for low stringency
conditions). Medium
stringency conditions include and encompass from at least about 16% v/v to at
least about
30% v/v formamide and from at least about 0.5 M to at least about 0.9 M salt
for
hybridization at 42 C, and at least about 0.1 M to at least about 0.2 M salt
for washing at 55
C. Medium stringency conditions also may include 1% Bovine Serum Albumin
(BSA), 1
mM EDTA, 0.5 M NaHPO4 (pH 7.2), 7% SDS for hybridization at 65 C, and (i) 2 x
SSC,
0.1% SDS; or (ii) 0.5% BSA, 1 mM EDTA, 40 mM NaHPO4 (pH 7.2), 5% SDS for
washing
at 60-65 C. One embodiment of medium stringency conditions includes
hybridizing in 6 x
SSC at about 45 C, followed by one or more washes in 0.2 x SSC, 0.1% SDS at
60 C. High
stringency conditions include and encompass from at least about 31% v/v to at
least about
50% v/v formamide and from about 0.01 M to about 0.15 M salt for hybridization
at 42 C,
and about 0.01 M to about 0.02 M salt for washing at 55 C. High stringency
conditions also
may include 1% BSA, 1 mM EDTA, 0.5 M NaHPO4 (pH 7.2), 7% SDS for hybridization
at
65 C, and (i) 0.2 x SSC, 0.1% SDS; or (ii) 0.5% BSA, 1 mM EDTA, 40 mM NaHPO4
(pH
7.2), 1% SDS for washing at a temperature in excess of 65 C. One embodiment
of high
stringency conditions includes hybridizing in 6 x SSC at about 45 C, followed
by one or
more washes in 0.2 x SSC, 0.1% SDS at 65 C.
[0480] In certain embodiments, a corresponding IRS biomarker polynucleotide is
one that
hybridizes to a disclosed nucleotide sequence under very high stringency
conditions. One
embodiment of very high stringency conditions includes hybridizing 0.5 M
sodium
phosphate, 7% SDS at 65 C, followed by one or more washes at 0.2 x SSC, 1%
SDS at 65
C.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 140 -
[0481] Other stringency conditions are well known in the art and a skilled
addressee will
recognize that various factors can be manipulated to optimize the specificity
of the
hybridization. Optimization of the stringency of the final washes can serve to
ensure a high
degree of hybridization. For detailed examples, see Ausubel et at., supra at
pages 2.10.1 to
2.10.16 and Sambrook et al. (1989, supra) at sections 1.101 to 1.104.
[0482] The IRS biomarkers disclosed herein each have significant sensitivity
and specificity
for diagnosing the presence, absence or degree of at least condition selected
from a healthy
condition (e.g., a normal condition or one in which inSIRS and inSIRS are
absent), inSIRS,
ipSIRS or a stage of ipSIRS (e.g., a stage of ipSIRS with a particular
severity such as mild
sepsis, severe sepsis and septic shock). Accordingly, it is feasible to use
individual IRS
biomarkers in methods, apparatus and kits that do not rely on the use of low
mutual
correlation between biomarkers to diagnose the presence, absence or degree of
the at least
condition. In illustrative examples of this type, the invention contemplates
methods, kits and
apparatus that are useful for diagnosing that inSIRS or a healthy condition is
present or
absent in a biological subject, which suitably involve: (1) correlating a
reference biomarker
signature with the presence or absence of a condition selected from inSIRS and
a healthy
condition, wherein the reference biomarker signature evaluates at least one
IRS biomarker;
(2) obtaining a biomarker signature of a sample from a subject, wherein the
sample
biomarker signature evaluates for an individual IRS biomarker in the reference
biomarker
signature a corresponding IRS biomarker; and (3) diagnosing the presence or
absence of the
condition in the subject based on the sample biomarker signature and the
reference biomarker
signature, wherein an individual IRS biomarker is an expression product of an
IRS biomarker
gene selected from the group A and group B IRS biomarker genes as defined
herein.
[0483] In other non-limiting examples, the methods, apparatus and kits are
useful for
diagnosing that ipSIRS or a healthy condition is present or absent in the
biological subject,
which suitably involve: (1) correlating a reference biomarker signature with
the presence or
absence of a condition selected from ipSIRS and a healthy condition, wherein
the reference
biomarker signature evaluates at least one IRS biomarker; (2) obtaining a
biomarker
signature of a sample from a subject, wherein the sample biomarker signature
evaluates for
an individual IRS biomarker in the reference biomarker signature a
corresponding IRS
biomarker; and (3) diagnosing the presence or absence of the condition in the
subject based

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 141 -
on the sample biomarker signature and the reference biomarker signature,
wherein an
individual IRS biomarker is an expression product of an IRS biomarker gene
selected from
the group C and group D IRS biomarker genes as defined herein.
[0484] In still other non-limiting examples, the methods, apparatus and kits
are useful for
diagnosing that inSIRS or ipSIRS is present or absent in the biological
subject, which
suitably involve: (1) correlating a reference biomarker signature with the
presence or absence
of a condition selected from inSIRS and ipSIRS, wherein the reference
biomarker signature
evaluates at least one IRS biomarker; (2) obtaining a biomarker signature of a
sample from a
subject, wherein the sample biomarker signature evaluates for an individual
IRS biomarker in
the reference biomarker signature a corresponding IRS biomarker; and (3)
diagnosing the
presence or absence of the condition in the subject based on the sample
biomarker signature
and the reference biomarker signature, wherein an individual IRS biomarker is
an expression
product of an IRS biomarker gene selected from the group E and group F IRS
biomarker
genes as defined herein.
[0485] In still other illustrative examples, the methods, apparatus and kits
are useful for
diagnosing that mild sepsis or severe sepsis is present or absent in the
biological subject,
which suitably involve: (1) correlating a reference biomarker signature with
the presence or
absence of a condition selected from mild sepsis and severe sepsis, wherein
the reference
biomarker signature evaluates at least one IRS biomarker; (2) obtaining a
biomarker
signature of a sample from a subject, wherein the sample biomarker signature
evaluates for
an individual IRS biomarker in the reference biomarker signature a
corresponding IRS
biomarker; and (3) diagnosing the presence or absence of the condition in the
subject based
on the sample biomarker signature and the reference biomarker signature,
wherein an
individual IRS biomarker is an expression product of an IRS biomarker gene
selected from
the group K and group L IRS biomarker genes as defined herein.
[0486] In still other illustrative examples, the methods, apparatus and kits
are useful for
diagnosing that mild sepsis or septic shock is present or absent in the
biological subject,
which suitably involve: (1) correlating a reference biomarker signature with
the presence or
absence of a condition selected from mild sepsis and septic shock, wherein the
reference
biomarker signature evaluates at least one IRS biomarker; (2) obtaining a
biomarker
signature of a sample from a subject, wherein the sample biomarker signature
evaluates for

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 142 -
an individual IRS biomarker in the reference biomarker signature a
corresponding IRS
biomarker; and (3) diagnosing the presence or absence of the condition in the
subject based
on the sample biomarker signature and the reference biomarker signature,
wherein an
individual IRS biomarker is an expression product of an IRS biomarker gene
selected from
the group M and group N IRS biomarker genes as defined herein.
[0487] In other non-limiting examples, the methods, apparatus and kits are
useful for
diagnosing that severe sepsis or septic shock is present or absent in the
biological subject,
which suitably involve: (1) correlating a reference biomarker signature with
the presence or
absence of a condition selected from severe sepsis and septic shock, wherein
the reference
biomarker signature evaluates at least one IRS biomarker; (2) obtaining a
biomarker
signature of a sample from a subject, wherein the sample biomarker signature
evaluates for
an individual IRS biomarker in the reference biomarker signature a
corresponding IRS
biomarker; and (3) diagnosing the presence or absence of the condition in the
subject based
on the sample biomarker signature and the reference biomarker signature,
wherein an
individual IRS biomarker is an expression product of an IRS biomarker gene
selected from
the group 0 and group P IRS biomarker genes as defined herein.
[0488] The biomarkers may be quantified or detected using any suitable
technique. In
specific embodiments, the biomarkers, including the IRS biomarkers, are
quantified using
reagents that determine the level or abundance of individual biomarkers. Non-
limiting
reagents of this type include reagents for use in nucleic acid- and protein-
based assays.
[0489] In illustrative nucleic acid-based assays, nucleic acid is isolated
from cells contained
in the biological sample according to standard methodologies (Sambrook, et
at., 1989, supra;
and Ausubel et at., 1994, supra). The nucleic acid is typically fractionated
(e.g., poly A+
RNA) or whole cell RNA. Where RNA is used as the subject of detection, it may
be desired
to convert the RNA to a complementary DNA. In some embodiments, the nucleic
acid is
amplified by a template-dependent nucleic acid amplification technique. A
number of
template dependent processes are available to amplify the IRS biomarker
sequences present
in a given template sample. An exemplary nucleic acid amplification technique
is the
polymerase chain reaction (referred to as PCR), which is described in detail
in U.S. Pat. Nos.
4,683,195, 4,683,202 and 4,800,159, Ausubel et at. (supra), and in Innis et
at., ("PCR
Protocols", Academic Press, Inc., San Diego Calif., 1990). Briefly, in PCR,
two primer

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 143 -
sequences are prepared that are complementary to regions on opposite
complementary
strands of the biomarker sequence. An excess of deoxynucleotide triphosphates
are added to
a reaction mixture along with a DNA polymerase, e.g., Taq polymerase. If a
cognate IRS
biomarker sequence is present in a sample, the primers will bind to the
biomarker and the
polymerase will cause the primers to be extended along the biomarker sequence
by adding on
nucleotides. By raising and lowering the temperature of the reaction mixture,
the extended
primers will dissociate from the biomarker to form reaction products, excess
primers will
bind to the biomarker and to the reaction products and the process is
repeated. A reverse
transcriptase PCR amplification procedure may be performed in order to
quantify the amount
of mRNA amplified. Methods of reverse transcribing RNA into cDNA are well
known and
described in Sambrook et at., 1989, supra. Alternative methods for reverse
transcription
utilize thermostable, RNA-dependent DNA polymerases. These methods are
described in
WO 90/07641. Polymerase chain reaction methodologies are well known in the
art.
[0490] In certain advantageous embodiments, the template-dependent
amplification involves
quantification of transcripts in real-time. For example, RNA or DNA may be
quantified using
the Real-Time PCR technique (Higuchi, 1992, et at., Biotechnology 10: 413-
417). By
determining the concentration of the amplified products of the target DNA in
PCR reactions
that have completed the same number of cycles and are in their linear ranges,
it is possible to
determine the relative concentrations of the specific target sequence in the
original DNA
mixture. If the DNA mixtures are cDNAs synthesized from RNAs isolated from
different
tissues or cells, the relative abundance of the specific mRNA from which the
target sequence
was derived can be determined for the respective tissues or cells. This direct
proportionality
between the concentration of the PCR products and the relative mRNA abundance
is only
true in the linear range of the PCR reaction. The final concentration of the
target DNA in the
plateau portion of the curve is determined by the availability of reagents in
the reaction mix
and is independent of the original concentration of target DNA. In specific
embodiments,
multiplexed, tandem PCR (MT-PCR) is employed, which uses a two-step process
for gene
expression profiling from small quantities of RNA or DNA, as described for
example in US
Pat. Appl. Pub. No. 20070190540. In the first step, RNA is converted into cDNA
and
amplified using multiplexed gene specific primers. In the second step each
individual gene is
quantitated by real time PCR.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 144 -
[0491] In certain embodiments, target nucleic acids are quantified using
blotting techniques,
which are well known to those of skill in the art. Southern blotting involves
the use of DNA
as a target, whereas Northern blotting involves the use of RNA as a target.
Each provides
different types of information, although cDNA blotting is analogous, in many
aspects, to
blotting or RNA species. Briefly, a probe is used to target a DNA or RNA
species that has
been immobilized on a suitable matrix, often a filter of nitrocellulose. The
different species
should be spatially separated to facilitate analysis. This often is
accomplished by gel
electrophoresis of nucleic acid species followed by "blotting" on to the
filter. Subsequently,
the blotted target is incubated with a probe (usually labeled) under
conditions that promote
denaturation and rehybridization. Because the probe is designed to base pair
with the target,
the probe will bind a portion of the target sequence under renaturing
conditions. Unbound
probe is then removed, and detection is accomplished as described above.
Following
detection/quantification, one may compare the results seen in a given subject
with a control
reaction or a statistically significant reference group or population of
control subjects as
defined herein. In this way, it is possible to correlate the amount of an IRS
biomarker nucleic
acid detected with the progression or severity of the disease. As used herein,
the term "probe"
refers to a molecule that binds to a specific sequence or sub-sequence or
other moiety of
another molecule. Unless otherwise indicated, the term "probe" typically
refers to a nucleic
acid probe that binds to another nucleic acid, also referred to herein as a
"target
polynucleotide", through complementary base pairing. Probes can bind target
polynucleotides lacking complete sequence complementarity with the probe,
depending on
the stringency of the hybridization conditions. Probes can be labeled directly
or indirectly and
include primers within their scope. By "primer" is meant an oligonucleotide
which, when
paired with a strand of DNA, is capable of initiating the synthesis of a
primer extension
product in the presence of a suitable polymerizing agent. The primer is
preferably single-
stranded for maximum efficiency in amplification but can alternatively be
double-stranded. A
primer must be sufficiently long to prime the synthesis of extension products
in the presence
of the polymerization agent. The length of the primer depends on many factors,
including
application, temperature to be employed, template reaction conditions, other
reagents, and
source of primers. For example, depending on the complexity of the target
sequence, the
primer may be at least about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17,
18, 19, 20, 21, 22, 23,
24, 25, 26, 27, 28, 29, 30, 35, 40, 50, 75, 100, 150, 200, 300, 400, 500, to
one base shorter in

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 145 -
length than the template sequence at the 3' end of the primer to allow
extension of a nucleic
acid chain, though the 5' end of the primer may extend in length beyond the 3'
end of the
template sequence. In certain embodiments, primers can be large
polynucleotides, such as
from about 35 nucleotides to several kilobases or more. Primers can be
selected to be
"substantially complementary" to the sequence on the template to which it is
designed to
hybridize and serve as a site for the initiation of synthesis. By
"substantially
complementary", it is meant that the primer is sufficiently complementary to
hybridize with a
target polynucleotide. Desirably, the primer contains no mismatches with the
template to
which it is designed to hybridize but this is not essential. For example, non-
complementary
nucleotide residues can be attached to the 5' end of the primer, with the
remainder of the
primer sequence being complementary to the template. Alternatively, non-
complementary
nucleotide residues or a stretch of non-complementary nucleotide residues can
be
interspersed into a primer, provided that the primer sequence has sufficient
complementarity
with the sequence of the template to hybridize therewith and thereby form a
template for
synthesis of the extension product of the primer.
[0492] Also contemplated are biochip-based technologies such as those
described by Hacia
et at. (1996, Nature Genetics 14: 441-447) and Shoemaker et at. (1996, Nature
Genetics 14:
450-456). Briefly, these techniques involve quantitative methods for analyzing
large numbers
of genes rapidly and accurately. By tagging genes with oligonucleotides or
using fixed
nucleic acid probe arrays, one can employ biochip technology to segregate
target molecules
as high-density arrays and screen these molecules on the basis of
hybridization. See also
Pease et at. (1994, Proc. Natl. Acad. Sci. U.S.A. 91: 5022-5026); Fodor et at.
(1991, Science
251: 767-773). Briefly, nucleic acid probes to IRS biomarker polynucleotides
are made and
attached to biochips to be used in screening and diagnostic methods, as
outlined herein. The
nucleic acid probes attached to the biochip are designed to be substantially
complementary to
specific expressed IRS biomarker nucleic acids, i.e., the target sequence
(either the target
sequence of the sample or to other probe sequences, for example in sandwich
assays), such
that hybridization of the target sequence and the probes of the present
invention occur. This
complementarity need not be perfect; there may be any number of base pair
mismatches,
which will interfere with hybridization between the target sequence and the
nucleic acid
probes of the present invention. However, if the number of mismatches is so
great that no
hybridization can occur under even the least stringent of hybridization
conditions, the

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 146 -
sequence is not a complementary target sequence. In certain embodiments, more
than one
probe per sequence is used, with either overlapping probes or probes to
different sections of
the target being used. That is, two, three, four or more probes, with three
being desirable, are
used to build in a redundancy for a particular target. The probes can be
overlapping (i.e. have
some sequence in common), or separate.
[0493] In an illustrative biochip analysis, oligonucleotide probes on the
biochip are exposed
to or contacted with a nucleic acid sample suspected of containing one or more
IRS
biomarker polynucleotides under conditions favoring specific hybridization.
Sample extracts
of DNA or RNA, either single or double-stranded, may be prepared from fluid
suspensions of
biological materials, or by grinding biological materials, or following a cell
lysis step which
includes, but is not limited to, lysis effected by treatment with SDS (or
other detergents),
osmotic shock, guanidinium isothiocyanate and lysozyme. Suitable DNA, which
may be used
in the method of the invention, includes cDNA. Such DNA may be prepared by any
one of a
number of commonly used protocols as for example described in Ausubel, et at.,
1994,
supra, and Sambrook, et al., 1989, supra.
[0494] Suitable RNA, which may be used in the method of the invention,
includes messenger
RNA, complementary RNA transcribed from DNA (cRNA) or genomic or subgenomic
RNA.
Such RNA may be prepared using standard protocols as for example described in
the relevant
sections of Ausubel, et al. 1994, supra and Sambrook, et al. 1989, supra).
[0495] cDNA may be fragmented, for example, by sonication or by treatment with
restriction
endonucleases. Suitably, cDNA is fragmented such that resultant DNA fragments
are of a
length greater than the length of the immobilized oligonucleotide probe(s) but
small enough
to allow rapid access thereto under suitable hybridization conditions.
Alternatively, fragments
of cDNA may be selected and amplified using a suitable nucleotide
amplification technique,
as described for example above, involving appropriate random or specific
primers.
[0496] Usually the target IRS biomarker polynucleotides are detectably labeled
so that their
hybridization to individual probes can be determined. The target
polynucleotides are typically
detectably labeled with a reporter molecule illustrative examples of which
include
chromogens, catalysts, enzymes, fluorochromes, chemiluminescent molecules,
bioluminescent molecules, lanthanide ions (e.g., Eu34), a radioisotope and a
direct visual
label. In the case of a direct visual label, use may be made of a colloidal
metallic or non-

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 147 -
metallic particle, a dye particle, an enzyme or a substrate, an organic
polymer, a latex
particle, a liposome, or other vesicle containing a signal producing substance
and the like.
Illustrative labels of this type include large colloids, for example, metal
colloids such as those
from gold, selenium, silver, tin and titanium oxide. In some embodiments in
which an
enzyme is used as a direct visual label, biotinylated bases are incorporated
into a target
polynucleotide.
[0497] The hybrid-forming step can be performed under suitable conditions for
hybridizing
oligonucleotide probes to test nucleic acid including DNA or RNA. In this
regard, reference
may be made, for example, to NUCLEIC ACID HYBRIDIZATION, A PRACTICAL
APPROACH (Homes and Higgins, eds.) (IRL press, Washington D.C., 1985). In
general,
whether hybridization takes place is influenced by the length of the
oligonucleotide probe
and the polynucleotide sequence under test, the pH, the temperature, the
concentration of
mono- and divalent cations, the proportion of G and C nucleotides in the
hybrid-forming
region, the viscosity of the medium and the possible presence of denaturants.
Such variables
also influence the time required for hybridization. The preferred conditions
will therefore
depend upon the particular application. Such empirical conditions, however,
can be routinely
determined without undue experimentation.
[0498] After the hybrid-forming step, the probes are washed to remove any
unbound nucleic
acid with a hybridization buffer. This washing step leaves only bound target
polynucleotides.
The probes are then examined to identify which probes have hybridized to a
target
polynucleotide.
[0499] The hybridization reactions are then detected to determine which of the
probes has
hybridized to a corresponding target sequence. Depending on the nature of the
reporter
molecule associated with a target polynucleotide, a signal may be
instrumentally detected by
irradiating a fluorescent label with light and detecting fluorescence in a
fluorimeter; by
providing for an enzyme system to produce a dye which could be detected using
a
spectrophotometer; or detection of a dye particle or a colored colloidal
metallic or non-
metallic particle using a reflectometer; in the case of using a radioactive
label or
chemiluminescent molecule employing a radiation counter or autoradiography.
Accordingly,
a detection means may be adapted to detect or scan light associated with the
label which light
may include fluorescent, luminescent, focused beam or laser light. In such a
case, a charge

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 148 -
couple device (CCD) or a photocell can be used to scan for emission of light
from a
probe:target polynucleotide hybrid from each location in the micro-array and
record the data
directly in a digital computer. In some cases, electronic detection of the
signal may not be
necessary. For example, with enzymatically generated color spots associated
with nucleic
acid array format, visual examination of the array will allow interpretation
of the pattern on
the array. In the case of a nucleic acid array, the detection means is
suitably interfaced with
pattern recognition software to convert the pattern of signals from the array
into a plain
language genetic profile. In certain embodiments, oligonucleotide probes
specific for
different IRS biomarker polynucleotides are in the form of a nucleic acid
array and detection
of a signal generated from a reporter molecule on the array is performed using
a 'chip
reader'. A detection system that can be used by a 'chip reader' is described
for example by
Pirrung et at. (U.S. Patent No. 5,143,854). The chip reader will typically
also incorporate
some signal processing to determine whether the signal at a particular array
position or
feature is a true positive or maybe a spurious signal. Exemplary chip readers
are described for
example by Fodor et at. (U.S. Patent No., 5,925,525). Alternatively, when the
array is made
using a mixture of individually addressable kinds of labeled microbeads, the
reaction may be
detected using flow cytometry.
[0500] In certain embodiments, the IRS biomarker is a target RNA (e.g., mRNA)
or a DNA
copy of the target RNA whose level is measured using at least one nucleic acid
probe that
hybridizes under at least low, medium, or high stringency conditions to the
target RNA or to
the DNA copy, wherein the nucleic acid probe comprises at least 15 (e.g., 15,
16, 17, 18, 19,
20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more) contiguous nucleotides of
an IRS
biomarker polynucleotide. In some embodiments, the measured level or abundance
of the
target RNA or its DNA copy is normalized to the level or abundance of a
reference RNA or a
DNA copy of the reference RNA. Suitably, the nucleic acid probe is immobilized
on a solid
or semi-solid support. In illustrative examples of this type, the nucleic acid
probe forms part
of a spatial array of nucleic acid probes. In some embodiments, the level of
nucleic acid
probe that is bound to the target RNA or to the DNA copy is measured by
hybridization (e.g.,
using a nucleic acid array). In other embodiments, the level of nucleic acid
probe that is
bound to the target RNA or to the DNA copy is measured by nucleic acid
amplification (e.g.,
using a polymerase chain reaction (PCR)). In still other embodiments, the
level of nucleic

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 149 -
acid probe that is bound to the target RNA or to the DNA copy is measured by
nuclease
protection assay.
[0501] In other embodiments, IRS biomarker protein levels are assayed using
protein-based
assays known in the art. For example, when an IRS biomarker protein is an
enzyme, the
protein can be quantified based upon its catalytic activity or based upon the
number of
molecules of the protein contained in a sample. Antibody-based techniques may
be employed
including, for example, immunoassays, such as the enzyme-linked immunosorbent
assay
(ELISA) and the radioimmunoassay (RIA).
[0502] In specific embodiments, protein-capture arrays that permit
simultaneous detection
and/or quantification of a large number of proteins are employed. For example,
low-density
protein arrays on filter membranes, such as the universal protein array system
(Ge, 2000
Nucleic Acids Res. 28(2):e3) allow imaging of arrayed antigens using standard
ELISA
techniques and a scanning charge-coupled device (CCD) detector. Immuno-sensor
arrays
have also been developed that enable the simultaneous detection of clinical
analytes. It is
now possible using protein arrays, to profile protein expression in bodily
fluids, such as in
sera of healthy or diseased subjects, as well as in subjects pre- and post-
drug treatment.
[0503] Exemplary protein capture arrays include arrays comprising spatially
addressed
antigen-binding molecules, commonly referred to as antibody arrays, which can
facilitate
extensive parallel analysis of numerous proteins defining a proteome or
subproteome.
Antibody arrays have been shown to have the required properties of specificity
and
acceptable background, and some are available commercially (e.g., BD
Biosciences,
Clontech, Bio-Rad and Sigma). Various methods for the preparation of antibody
arrays have
been reported (see, e.g., Lopez et al., 2003 1 Chromatogram. B 787:19-27;
Cahill, 2000
Trends in Biotechnology 7:47-51; U.S. Pat. App. Pub. 2002/0055186; U.S. Pat.
App. Pub.
2003/0003599; PCT publication WO 03/062444; PCT publication WO 03/077851; PCT
publication WO 02/59601; PCT publication WO 02/39120; PCT publication WO
01/79849;
PCT publication WO 99/39210). The antigen-binding molecules of such arrays may

recognize at least a subset of proteins expressed by a cell or population of
cells, illustrative
examples of which include growth factor receptors, hormone receptors,
neurotransmitter
receptors, catecholamine receptors, amino acid derivative receptors, cytokine
receptors,
extracellular matrix receptors, antibodies, lectins, cytokines, serpins,
proteases, kinases,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 150 -
phosphatases, ras-like GTPases, hydrolases, steroid hormone receptors,
transcription factors,
heat-shock transcription factors, DNA-binding proteins, zinc-finger proteins,
leucine-zipper
proteins, homeodomain proteins, intracellular signal transduction modulators
and effectors,
apoptosis-related factors, DNA synthesis factors, DNA repair factors, DNA
recombination
factors and cell-surface antigens.
[0504] Individual spatially distinct protein-capture agents are typically
attached to a support
surface, which is generally planar or contoured. Common physical supports
include glass
slides, silicon, microwells, nitrocellulose or PVDF membranes, and magnetic
and other
microbeads.
[0505] Particles in suspension can also be used as the basis of arrays,
providing they are
coded for identification; systems include color coding for microbeads (e.g.,
available from
Luminex, Bio-Rad and Nanomics Biosystems) and semiconductor nanocrystals
(e.g.,
QD0t5TM, available from Quantum Dots), and barcoding for beads (UltraPlexTM,
available
from Smartbeads) and multimetal microrods (NanobarcodesTM particles, available
from
Surromed). Beads can also be assembled into planar arrays on semiconductor
chips (e.g.,
available from LEAPS technology and BioArray Solutions). Where particles are
used,
individual protein-capture agents are typically attached to an individual
particle to provide
the spatial definition or separation of the array. The particles may then be
assayed separately,
but in parallel, in a compartmentalized way, for example in the wells of a
microtiter plate or
in separate test tubes.
[0506] In operation, a protein sample, which is optionally fragmented to form
peptide
fragments (see, e.g., U.S. Pat. App. Pub. 2002/0055186), is delivered to a
protein-capture
array under conditions suitable for protein or peptide binding, and the array
is washed to
remove unbound or non-specifically bound components of the sample from the
array. Next,
the presence or amount of protein or peptide bound to each feature of the
array is detected
using a suitable detection system. The amount of protein bound to a feature of
the array may
be determined relative to the amount of a second protein bound to a second
feature of the
array. In certain embodiments, the amount of the second protein in the sample
is already
known or known to be invariant.
[0507] In specific embodiments, the IRS biomarker is a target polypeptide
whose level is
measured using at least one antigen-binding molecule that is immuno-
interactive with the

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 151 -
target polypeptide. In these embodiments, the measured level of the target
polypeptide is
normalized to the level of a reference polypeptide. Suitably, the antigen-
binding molecule is
immobilized on a solid or semi-solid support. In illustrative examples of this
type, the
antigen-binding molecule forms part of a spatial array of antigen-binding
molecule. In some
embodiments, the level of antigen-binding molecule that is bound to the target
polypeptide is
measured by immunoassay (e.g., using an ELISA). Reference herein to "immuno-
interactive"
includes reference to any interaction, reaction, or other form of association
between
molecules and in particular where one of the molecules is, or mimics, a
component of the
immune system.
[0508] All the essential reagents required for detecting and quantifying the
biomarkers of the
invention, including IRS biomarkers, may be assembled together in a kit. In
some
embodiments, the kit comprises: (i) a reagent that allows quantification
(e.g., determining the
level or abundance) of a first biomarker; and (ii) a reagent that allows
quantification (e.g.,
determining the level or abundance) of a second biomarker, wherein the first
and second
biomarkers have a mutual correlation in respect of at least one condition
(e.g., at least one of
a healthy condition and one or more diseases such as but not limited to
inSIRS, ipSIRS or a
stage of ipSIRS (e.g., a stage of ipSIRS with a particular severity such as
mild sepsis, severe
sepsis and septic shock)) that lies within a mutual correlation range of
between 0.9, and
wherein a combination of respective biomarker values for the first and second
biomarkers
that are measured for or derived from a biological subject has a performance
value greater
than or equal to a performance threshold representing the ability of the
combination of the
first and second biomarkers to diagnose the presence, absence or degree of the
at least one
condition, or to provide a prognosis for the at least one condition, the
performance threshold
being a variance explained of at least 0.3. In some embodiments, the kit
further comprises
(iii) a reagent that allows quantification (e.g., determining the level or
abundance) of a third
biomarker; and (iv) a reagent that allows quantification (e.g., determining
the level or
abundance) of a fourth biomarker, wherein the third and fourth biomarkers have
a mutual
correlation in respect of at the least one condition that lies within a mutual
correlation range
of between 0.9, and wherein a combination of respective biomarker values for
the third and
fourth biomarkers that are measured for or derived from a biological subject
has a
performance value greater than or equal to a performance threshold
representing the ability of
the combination of the third and fourth biomarkers to diagnose the presence,
absence or

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 152 -
degree of the at least one condition, or to provide a prognosis for the at
least one condition,
the performance threshold being a variance explained of at least 0.3.
[0509] In advantageous embodiments, the kits of the present invention are
useful for
diagnosing the presence, absence or degree of at least one condition, or for
providing a
prognosis for at least one condition, wherein the at least one condition is
selected from a
healthy condition, inSIRS, ipSIRS or a stage of ipSIRS. In these embodiments,
IRS
biomarkers are suitably selected from a group as broadly described above and
elsewhere
herein.
[0510] In the context of the present invention, "kit" is understood to mean a
product
containing the different reagents necessary for carrying out the methods of
the invention
packed so as to allow their transport and storage. Materials suitable for
packing the
components of the kit include crystal, plastic (polyethylene, polypropylene,
polycarbonate
and the like), bottles, vials, paper, envelopes and the like. Additionally,
the kits of the
invention can contain instructions for the simultaneous, sequential or
separate use of the
different components contained in the kit. The instructions can be in the form
of printed
material or in the form of an electronic support capable of storing
instructions such that they
can be read by a subject, such as electronic storage media (magnetic disks,
tapes and the
like), optical media (CD-ROM, DVD) and the like. Alternatively or in addition,
the media
can contain Internet addresses that provide the instructions.
[0511] A "reagent that allows quantification of a biomarker" means a compound
or material,
or set of compounds or materials, which allow quantification of the biomarker.
In specific
embodiments, the compound, material or set of compounds or materials permit
determining
the expression level of a gene (e.g., an IRS biomarker gene), including
without limitation the
extraction of RNA material, the determination of the level of a corresponding
RNA, etc.,
primers for the synthesis of a corresponding cDNA, primers for amplification
of DNA,
and/or probes capable of specifically hybridizing with the RNAs (or the
corresponding
cDNAs) encoded by the genes, TaqMan probes, etc.
[0512] The kits may also optionally include appropriate reagents for detection
of labels,
positive and negative controls, washing solutions, blotting membranes,
microtiter plates,
dilution buffers and the like. For example, a nucleic acid-based detection kit
may include (i) a
biomarker polynucleotide (e.g., an IRS biomarker polynucleotide) (which may be
used as a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 153 -
positive control), (ii) a primer or probe that specifically hybridizes to a
biomarker
polynucleotide (e.g., an IRS biomarker polynucleotide). Also included may be
enzymes
suitable for amplifying nucleic acids including various polymerases (reverse
transcriptase,
Taq, SequenaseTM, DNA ligase etc. depending on the nucleic acid amplification
technique
employed), deoxynucleotides and buffers to provide the necessary reaction
mixture for
amplification. Such kits also generally will comprise, in suitable means,
distinct containers
for each individual reagent and enzyme as well as for each primer or probe.
Alternatively, a
protein-based detection kit may include (i) a biomarker polypeptide (e.g., an
IRS biomarker
polypeptide) (which may be used as a positive control), (ii) an antibody that
binds
specifically to a biomarker polypeptide (e.g., an IRS biomarker polypeptide).
The kit can also
feature various devices (e.g., one or more) and reagents (e.g., one or more)
for performing
one of the assays described herein; and/or printed instructions for using the
kit to quantify the
expression of a biomarker gene (e.g., an IRS biomarker gene).
[0513] The reagents described herein, which may be optionally associated with
detectable
labels, can be presented in the format of a microfluidics card, a chip or
chamber, a microarray
or a kit adapted for use with the assays described in the examples or below,
e.g., RT-PCR or
Q PCR techniques described herein. The term "microarray" refers to an
arrangement of
hybridizable array elements, e.g., probes (including primers), ligands,
biomarker nucleic acid
sequence or protein sequences on a substrate.
[0514] The reagents also have utility in compositions for detecting and
quantifying the
biomarkers of the invention. For example, a reverse transcriptase may be used
to reverse
transcribe RNA transcripts, including mRNA, in a nucleic acid sample, to
produce reverse
transcribed transcripts, including reverse transcribed mRNA (also referred to
as "cDNA").
The nucleic acid sample is suitably derived from components of the immune
system,
representative examples of which include components of the innate and adaptive
immune
systems as broadly discussed for example above. In specific embodiments, the
reverse
transcribed RNA is derived blood cells (e.g., peripheral blood cells).
Suitably, the reverse
transcribed RNA is derived leukocytes.
[0515] The reagents are suitably used to quantify the reverse transcribed
transcripts. For
example, oligonucleotide primers that hybridize to the reverse transcribed
transcript can be
used to amplify at least a portion of the reverse transcribed transcript via a
suitable nucleic

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 154 -
acid amplification technique, e.g., RT-PCR or Q PCR techniques described
herein.
Alternatively, oligonucleotide probes may be used to hybridize to the reverse
transcribed
transcript for the quantification, using a nucleic acid hybridization analysis
technique (e.g.,
microarray analysis), as described for example above. Thus, in some
embodiments, a
respective oligonucleotide primer or probe is hybridized to a complementary
nucleic acid
sequence of a reverse transcribed transcript in the compositions of the
invention. The
compositions typically comprise labeled reagents for detecting and/or
quantifying the reverse
transcribed transcripts. Representative reagents of this type include labeled
oligonucleotide
primers or probes that hybridize to RNA transcripts or reverse transcribed
RNA, labeled
RNA, labeled reverse transcribed RNA as well as labeled oligonucleotide
linkers or tags
(e.g., a labeled RNA or DNA linker or tag) for labeling (e.g., end labeling
such as 3' end
labeling) RNA or reverse transcribed RNA. The primers, probes, RNA or reverse
transcribed
RNA (i.e., cDNA) (whether labeled or non-labeled) may be immobilized or free
in solution.
Representative reagents of this type include labeled oligonucleotide primers
or probes that
hybridize to reverse transcribed and transcripts as well as labeled reverse
transcribed
transcripts. The label can be any reporter molecule as known in the art,
illustrative examples
of which are described above and elsewhere herein.
[0516] The present invention also encompasses non-reverse transcribed RNA
embodiments
in which cDNA is not made and the RNA transcripts are directly the subject of
the analysis.
Thus, in other embodiments, reagents are suitably used to quantify RNA
transcripts directly.
For example, oligonucleotide probes can be used to hybridize to transcripts
for quantification
of immune system biomarkers of the invention, using a nucleic acid
hybridization analysis
technique (e.g., microarray analysis), as described for example above. Thus,
in some
embodiments, a respective oligonucleotide probe is hybridized to a
complementary nucleic
acid sequence of an immune system biomarker transcript in the compositions of
the
invention. In illustrative examples of this type, the compositions may
comprise labeled
reagents that hybridize to transcripts for detecting and/or quantifying the
transcripts.
Representative reagents of this type include labeled oligonucleotide probes
that hybridize to
transcripts as well as labeled transcripts. The primers or probes may be
immobilized or free
in solution.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 155 -
[0517] The term "immobilized" means that a molecular species of interest is
fixed to a solid
support, suitably by covalent linkage. This covalent linkage can be achieved
by different
means depending on the molecular nature of the molecular species. Moreover,
the molecular
species may be also fixed on the solid support by electrostatic forces,
hydrophobic or
hydrophilic interactions or Van-der-Waals forces. The above described physico-
chemical
interactions typically occur in interactions between molecules. In particular
embodiments, all
that is required is that the molecules (e.g., nucleic acids or polypeptides)
remain immobilized
or attached to a support under conditions in which it is intended to use the
support, for
example in applications requiring nucleic acid amplification and/or sequencing
or in in
antibody-binding assays. For example, oligonucleotides or primers are
immobilized such that
a 3' end is available for enzymatic extension and/or at least a portion of the
sequence is
capable of hybridizing to a complementary sequence. In some embodiments,
immobilization
can occur via hybridization to a surface attached primer, in which case the
immobilized
primer or oligonucleotide may be in the 3'-5' orientation. In other
embodiments,
immobilization can occur by means other than base-pairing hybridization, such
as the
covalent attachment.
[0518] The term "solid support" as used herein refers to a solid inert surface
or body to
which a molecular species, such as a nucleic acid and polypeptides can be
immobilized. Non-
limiting examples of solid supports include glass surfaces, plastic surfaces,
latex, dextran,
polystyrene surfaces, polypropylene surfaces, polyacrylamide gels, gold
surfaces, and silicon
wafers. In some embodiments, the solid supports are in the form of membranes,
chips or
particles. For example, the solid support may be a glass surface (e.g., a
planar surface of a
flow cell channel). In some embodiments, the solid support may comprise an
inert substrate
or matrix which has been "functionalized", such as by applying a layer or
coating of an
intermediate material comprising reactive groups which permit covalent
attachment to
molecules such as polynucleotides. By way of non-limiting example, such
supports can
include polyacrylamide hydrogels supported on an inert substrate such as
glass. The
molecules (e.g., polynucleotides) can be directly covalently attached to the
intermediate
material (e.g., a hydrogel) but the intermediate material can itself be non-
covalently attached
to the substrate or matrix (e.g., a glass substrate). The support can include
a plurality of
particles or beads each having a different attached molecular species.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 156 -
[0519] The present invention also extends to the management of inSIRS, ipSIRS
or particular
stages of ipSIRS, or prevention of further progression of inSIRS, ipSIRS or
particular stages
of ipSIRS (e.g., mild sepsis, severe sepsis and septic shock), or assessment
of the efficacy of
therapies in subjects following positive diagnosis for the presence of inSIRS,
ipSIRS or
particular stage of ipSIRS (e.g., mild sepsis, severe sepsis and septic shock)
in a subject. The
management of inSIRS or ipSIRS conditions is generally highly intensive and
can include
identification and amelioration of the underlying cause and aggressive use of
therapeutic
compounds such as, vasoactive compounds, antibiotics, steroids, antibodies to
endotoxin,
anti-tumor necrosis factor agents, recombinant protein C. In addition,
palliative therapies as
described for example in Cohen and Glauser (1991, Lancet 338: 736-739) aimed
at restoring
and protecting organ function can be used such as intravenous fluids and
oxygen and tight
glycemic control. Therapies for ipSIRS are reviewed in Healy (2002, Ann
Pharmacother.
36(4): 648-54) and Brindley (2005, CJEM. 7(4): 227) and Jenkins (2006, J Hosp
Med. 1(5):
285-295).
[0520] Typically, the therapeutic agents will be administered in
pharmaceutical (or
veterinary) compositions together with a pharmaceutically acceptable carrier
and in an
effective amount to achieve their intended purpose. The dose of active
compounds
administered to a subject should be sufficient to achieve a beneficial
response in the subject
over time such as a reduction in, or relief from, the symptoms of inSIRS,
ipSIRS or particular
stages of ipSIRS. The quantity of the pharmaceutically active compounds(s) to
be
administered may depend on the subject to be treated inclusive of the age,
sex, weight and
general health condition thereof. In this regard, precise amounts of the
active compound(s)
for administration will depend on the judgment of the practitioner. In
determining the
effective amount of the active compound(s) to be administered in the treatment
or prevention
of inSIRS, ipSIRS or particular stages of ipSIRS, the medical practitioner or
veterinarian may
evaluate severity of any symptom associated with the presence of inSIRS,
ipSIRS or
particular stages of ipSIRS including, inflammation, blood pressure anomaly,
tachycardia,
tachypnea fever, chills, vomiting, diarrhea, skin rash, headaches, confusion,
muscle aches,
seizures. In any event, those of skill in the art may readily determine
suitable dosages of the
therapeutic agents and suitable treatment regimens without undue
experimentation.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 157 -
[0521] The therapeutic agents may be administered in concert with adjunctive
(palliative)
therapies to increase oxygen supply to major organs, increase blood flow to
major organs
and/or to reduce the inflammatory response. Illustrative examples of such
adjunctive
therapies include non-steroidal-anti-inflammatory drugs (NSAIDs), intravenous
saline and
oxygen.
[0522] In specific embodiments of the present invention, the methods,
apparatus and kits
described above and elsewhere herein are contemplated for use in methods of
treating,
preventing or inhibiting the development of at least one condition selected
from inSIRS,
ipSIRS or a particular stage of ipSIRS (e.g., mild sepsis, severe sepsis or
septic shock) in a
subject. These methods (also referred to herein as "treatment methods")
generally comprise:
(a) determining a plurality of IRS biomarker values, each IRS biomarker value
being
indicative of a value measured or derived for at least one (e.g., 1, 2, 3, 4,
5, 6, 7, 8, 9, 10, or
more) IRS biomarker of a biological subject; (b) determining an indicator
using a
combination of the plurality of IRS biomarker values, the indicator being at
least partially
indicative of the presence, absence or degree of the at least one condition,
wherein: (i) at least
two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) IRS biomarkers have a mutual
correlation in
respect of the at least one condition that lies within a mutual correlation
range, the mutual
correlation range being between 0.9; and (ii) the indicator has a performance
value greater
than or equal to a performance threshold representing the ability of the
indicator to diagnose
the presence, absence or degree of the at least one condition, the performance
threshold being
indicative of an explained variance of at least 0.3; and (c) administering to
the subject, on the
basis that the indicator indicates the presence of inSIRS, an effective amount
of an agent that
treats or ameliorates the symptoms or reverses or inhibits the development of
inSIRS, or
administering to the subject, on the basis that the indicator indicates the
presence of ipSIRS
or a particular stage of ipSIRS, an effective amount of an agent that treats
or ameliorates the
symptoms or reverses or inhibits the development of ipSIRS or the particular
stage of
ipSIRS.
[0523] In advantageous embodiments, the treatment methods comprise: (1)
determining a
plurality of measured IRS biomarker values, each measured IRS biomarker value
being a
measured value of an IRS biomarker of the biological subject; and (2) applying
a function to
at least one of the measured IRS biomarker values to determine at least one
derived IRS

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 158 -
biomarker value, the at least one derived IRS biomarker value being indicative
of a value of a
corresponding derived IRS biomarker. The function suitably includes at least
one of: (a)
multiplying two IRS biomarker values; (b) dividing two IRS biomarker values;
(c) adding
two IRS biomarker values; (d) subtracting two IRS biomarker values; (e) a
weighted sum of
at least two IRS biomarker values; (f) a log sum of at least two IRS biomarker
values; and (g)
a sigmoidal function of at least two IRS biomarker values.
[0524] In some embodiments the methods, apparatus and kits of the present
invention are
used for monitoring, treatment and management of conditions that can lead to
inSIRS or
ipSIRS, illustrative examples of which include retained placenta, meningitis,
endometriosis,
shock, toxic shock (i.e., sequelae to tampon use), gastroenteritis,
appendicitis, ulcerative
colitis, Crohn's disease, inflammatory bowel disease, acid gut syndrome, liver
failure and
cirrhosis, failure of colostrum transfer in neonates, ischemia (in any organ),
bacteremia,
infections within body cavities such as the peritoneal, pericardial, thecal,
and pleural cavities,
burns, severe wounds, excessive exercise or stress, hemodialysis, conditions
involving
intolerable pain (e.g., pancreatitis, kidney stones), surgical operations, and
non-healing
lesions. In these embodiments, the methods or kits of the present invention
are typically used
at a frequency that is effective to monitor the early development of inSIRS,
ipSIRS or
particular stages of ipSIRS, to thereby enable early therapeutic intervention
and treatment of
that condition. In illustrative examples, the diagnostic methods or kits are
used at least at 1, 2,
3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or
24 hour intervals or at
least 1, 2, 3, 4, 5 or 6 day intervals, or at least weekly, fortnightly or
monthly.
[0525] The present invention can be practiced in the field of predictive
medicine for the
purpose of diagnosis or monitoring the presence or development of a condition
selected from
inSIRS, ipSIRS or a particular stage of ipSIRS in a subject, and/or monitoring
response to
therapy efficacy.
[0526] The biomarker signatures and corresponding indicators of the present
invention
further enable determination of endpoints in pharmacotranslational studies.
For example,
clinical trials can take many months or even years to establish the
pharmacological
parameters for a medicament to be used in treating or preventing inSIRS,
ipSIRS or a
particular stage of ipSIRS (e.g., mild sepsis, severe sepsis and septic
shock). However, these
parameters may be associated with a biomarker signature and corresponding
indicator of a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 159 -
health state (e.g., a healthy condition). Hence, the clinical trial can be
expedited by selecting
a treatment regimen (e.g., medicament and pharmaceutical parameters), which
results in a
biomarker signature associated with a desired health state (e.g., healthy
condition). This may
be determined for example by: a) determining a plurality of IRS biomarker
values, each IRS
biomarker value being indicative of a value measured or derived for at least
one (e.g., 1, 2, 3,
4, 5, 6, 7, 8, 9, 10, or more) IRS biomarker of a biological subject after
treatment with a
treatment regimen; (b) determining an indicator using a combination of the
plurality of IRS
biomarker values, the indicator being at least partially indicative of the
presence, absence or
degree of at least one condition selected from a healthy condition, inSIRS,
ipSIRS or a
particular stage of ipSIRS, wherein: (i) at least two (e.g., 2, 3, 4, 5, 6, 7,
8, 9, 10, or more)
IRS biomarkers have a mutual correlation in respect of the at least one
condition that lies
within a mutual correlation range, the mutual correlation range being between
0.9; and (ii)
the indicator has a performance value greater than or equal to a performance
threshold
representing the ability of the indicator to diagnose the presence, absence or
degree of the at
least one condition, or to provide a prognosis for the at least one condition,
the performance
threshold being indicative of an explained variance of at least 0.3, and (c)
determining that
the treatment regimen is effective for changing the health status of the
subject to the desired
health state (e.g., healthy condition) on the basis that the indicator
indicates the presence of a
healthy condition or the presence of a condition of a lower degree relative to
the degree of the
condition in the subject before treatment with the treatment regimen. As used
herein, the term
"degree" refers to the extent or stage of a condition. Thus, for example, mild
sepsis is a stage
or degree of sepsis that is lower than severe sepsis. Similarly, severe sepsis
is a stage or
degree of sepsis that is lower than septic shock. Accordingly, this aspect of
the present
invention advantageously provides methods of monitoring the efficacy of a
particular
treatment regimen in a subject (for example, in the context of a clinical
trial) already
diagnosed with a condition selected from inSIRS, ipSIRS or a particular stage
of ipSIRS.
These methods take advantage of measured or derived IRS biomarker values that
correlate
with treatment efficacy to determine, for example, whether measured or derived
IRS
biomarker values of a subject undergoing treatment partially or completely
normalize during
the course of or following therapy or otherwise shows changes associated with
responsiveness to the therapy.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 160 -
[0527] As used herein, the term "treatment regimen" refers to prophylactic
and/or therapeutic
(i.e., after onset of a specified condition) treatments, unless the context
specifically indicates
otherwise. The term "treatment regimen" encompasses natural substances and
pharmaceutical
agents (i.e., "drugs") as well as any other treatment regimen including but
not limited to
dietary treatments, physical therapy or exercise regimens, surgical
interventions, and
combinations thereof
[0528] Accordingly, the invention provides methods of correlating a biomarker
signature
with an effective treatment regimen for a condition selected from inSIRS,
ipSIRS or a
particular stage of ipSIRS (e.g., mild sepsis, severe sepsis and septic
shock), wherein the
methods generally comprise: (a) determining a biomarker signature defining a
combination
of at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) IRS biomarker
values corresponding to
values of at least two IRS biomarkers that can be measured for or derived from
a biological
subject having the condition and for whom an effective treatment has been
identified,
wherein: (i) the at least two IRS biomarkers have a mutual correlation in
respect of the
condition that lies within a mutual correlation range, the mutual correlation
range being
between 0.9; and (ii) the combination of at least two biomarker values has a
performance
value greater than or equal to a performance threshold representing the
ability of the
combination of at least two biomarker values to diagnose the presence, absence
or degree of
the condition, or to provide a prognosis for the condition, the performance
threshold being
indicative of an explained variance of at least 0.3; and (b) correlating the
biomarker signature
so determined with an effective treatment regimen for the condition. The term
"correlating"
generally refers to determining a relationship between one type of data with
another or with a
state. In specific embodiments, an indicator or biomarker signature is
correlated to a global
probability or a particular outcome, using receiver operating characteristic
(ROC) curves.
[0529] The invention further provides methods of determining whether a
treatment regimen
is effective for treating a subject with a condition selected from inSIRS,
ipSIRS or a
particular stage of ipSIRS (e.g., mild sepsis, severe sepsis and septic
shock). These methods
generally comprise: (a) determining a plurality of post-treatment IRS
biomarker values, each
post-treatment IRS biomarker value being indicative of a value measured or
derived for at
least one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) IRS biomarker of a
biological subject after
treatment with the treatment regimen; (b) determining a post-treatment
indicator using a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 161 -
combination of the plurality of post-treatment IRS biomarker values, the post-
treatment
indicator being at least partially indicative of the presence, absence or
degree of at least one
condition selected from a healthy condition, inSIRS, ipSIRS or a particular
stage of ipSIRS,
wherein: (i) at the least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) IRS
biomarkers have a
mutual correlation in respect of the at least one condition that lies within a
mutual correlation
range, the mutual correlation range being between 0.9; and (ii) the post-
treatment indicator
has a performance value greater than or equal to a performance threshold
representing the
ability of the post-treatment indicator to diagnose the presence, absence or
degree of the at
least one condition, the performance threshold being indicative of an
explained variance of at
least 0.3, wherein the post-treatment indicator indicates whether the
treatment regimen is
effective for treating the condition in the subject on the basis that post-
treatment indicator
indicates the presence of a healthy condition or the presence of a condition
of a lower degree
relative to the degree of the condition in the subject before treatment with
the treatment
regimen.
[0530] The invention can also be practiced to evaluate whether a subject is
responding (i.e., a
positive response) or not responding (i.e., a negative response) to a
treatment regimen or has
a side effect to a treatment regimen. This aspect of the invention provides
methods of
correlating a biomarker signature with a positive or negative response or a
side effect to a
treatment regimen, which generally comprise: (a) determining a biomarker
signature defining
a combination of at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) IRS
biomarker values
corresponding to values of at least two IRS biomarkers that can be measured
for or derived
from a biological subject following commencement of the treatment regimen,
wherein: (i) the
at least two IRS biomarkers have a mutual correlation in respect of at least
one condition
selected from a healthy condition, inSIRS, ipSIRS or a particular stage of
ipSIRS, which lies
within a mutual correlation range, the mutual correlation range being between
0.9; and (ii)
the combination of at least two biomarker values has a performance value
greater than or
equal to a performance threshold representing the ability of the combination
of at least two
biomarker values to diagnose the presence, absence or degree of the at least
one condition, or
to provide a prognosis for the at least one condition, the performance
threshold being
indicative of an explained variance of at least 0.3; and (b) correlating the
biomarker signature
so determined with a positive or negative response to the treatment regimen.
As used herein,
the term "positive response" means that the result of the treatment regimen
includes some

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 162 -
clinically significant benefit, such as the prevention, or reduction of
severity, of symptoms, or
a slowing of the progression of the condition. By contrast, the term "negative
response"
means that the treatment regimen provides no clinically significant benefit,
such as the
prevention, or reduction of severity, of symptoms, or increases the rate of
progression of the
condition.
[0531] The invention also encompasses methods of determining a positive or
negative
response to a treatment regimen and/or a side effect of a treatment regimen by
a subject with
a condition selected from inSIRS, ipSIRS or a particular stage of ipSIRS.
These methods
generally comprise: (a) correlating a reference biomarker signature with a
positive or
negative response or a side effect to the treatment regimen, wherein the
biomarker signature
defines a combination of at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or
more) IRS biomarker
values corresponding to values of at least two IRS biomarkers that are
measured for or
derived from a control biological subject or control group, wherein: (i) the
at least two IRS
biomarkers have a mutual correlation in respect of at least one condition
selected from a
healthy condition, inSIRS, ipSIRS or a particular stage of ipSIRS, which lies
within a mutual
correlation range, the mutual correlation range being between 0.9; and (ii)
the combination
of at least two biomarker values has a performance value greater than or equal
to a
performance threshold representing the ability of the combination of at least
two biomarker
values to diagnose the presence, absence or degree of the at least one
condition, or to provide
a prognosis for the at least one condition, the performance threshold being
indicative of an
explained variance of at least 0.3; (b) determining a sample biomarker
signature defining a
combination of at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) IRS
biomarker values
corresponding to values of at least two IRS biomarkers that are measured for
or derived from
a biological subject following commencement of the treatment regimen, wherein:
(i) the at
least two IRS biomarkers have a mutual correlation in respect of at least one
condition
selected from a healthy condition, inSIRS, ipSIRS or a particular stage of
ipSIRS, which lies
within a mutual correlation range, the mutual correlation range being between
0.9; and (ii)
the combination of at least two biomarker values has a performance value
greater than or
equal to a performance threshold representing the ability of the combination
of at least two
biomarker values to diagnose the presence, absence or degree of the at least
one condition, or
to provide a prognosis for the at least one condition, the performance
threshold being
indicative of an explained variance of at least 0.3; wherein the sample
biomarker signature

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 163 -
indicates whether the subject is responding positively or negatively to the
treatment regimen
and/or is developing a side effect from the treatment regimen, based on the
correlation of the
reference biomarker signature with the positive or negative response or side
effect to the
treatment regimen.
[0532] In related embodiments, the present invention further contemplates
methods of
determining a positive or negative response to a treatment regimen and/or a
side effect to a
treatment regimen by a biological subject. These methods generally comprise:
(a)
determining a sample biomarker signature defining a combination of at least
two (e.g., 2, 3,
4, 5, 6, 7, 8, 9, 10, or more) IRS biomarker values corresponding to values of
at least two IRS
biomarkers that are measured for or derived from a biological subject
following
commencement of the treatment regimen, wherein: (i) the at least two IRS
biomarkers have a
mutual correlation in respect of at least one condition selected from a
healthy condition,
inSIRS, ipSIRS or a particular stage of ipSIRS, which lies within a mutual
correlation range,
the mutual correlation range being between 0.9; and (ii) the combination of
at least two
biomarker values has a performance value greater than or equal to a
performance threshold
representing the ability of the combination of at least two biomarker values
to diagnose the
presence, absence or degree of the at least one condition, or to provide a
prognosis for the at
least one condition, the performance threshold being indicative of an
explained variance of at
least 0.3, wherein the sample biomarker signature is correlated with a
positive or negative
response to the treatment regimen and/or to a side effect from the treatment
regimen; and (b)
determining whether the subject is responding positively or negatively to the
treatment
regimen and/or is developing a side effect from the treatment regimen based on
the sample
biomarker signature.
[0533] This above methods can be practiced to identify responders or non-
responders
relatively early in the treatment process, i.e., before clinical
manifestations of efficacy. In this
way, the treatment regimen can optionally be discontinued, a different
treatment protocol can
be implemented and/or supplemental therapy can be administered. Thus, in some
embodiments, a sample IRS biomarker signature is obtained within about 2
hours, 4 hours, 6
hours, 12 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 1 week, 2 weeks, 3
weeks, 4 weeks, 6
weeks, 8 weeks, 10 weeks, 12 weeks, 4 months, six months or longer of
commencing
therapy.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 164 -
[0534] A number of non-limiting example signatures for use in diagnosing
respective
conditions will now be described. For the purpose of illustration, the above-
described
process was used to select biomarkers that provided the theoretical best
diagnostic
biomarkers, selected from combinations including measured and/or derived
biomarkers.
Following this, other measured and/or derived biomarkers were grouped based on
their
correlation to the best diagnostic biomarkers, with the ability of biomarkers
within these
groups to act as a diagnostic signature then being assessed.
[0535] The results set out in detail below highlight that as long as the above-
described
criteria are met, the resulting signatures provide the required discriminatory
ability for use in
diagnosing the presence, absence, degree or prognosis of at least one
condition in a biological
subj ect.
Signature Derivation
[0536] An illustrative process for the identification of mRNA biomarkers for
use in
diagnostic algorithms will now be described.
Summary
[0537] Peripheral blood samples were obtained from healthy controls and
patients
retrospectively diagnosed by a panel of physicians with either inSIRS or
ipSIRS (blood
culture positive). ipSIRS patients were further classified retrospectively
into "mild",
"severe" or "shock" based on clinical parameters. Total RNA from patient
samples was then
used in gene expression analysis (GeneChip and / or quantitative PCR (qPCR)).
Gene
expression data were analyzed using a variety of statistical approaches to
identify individual
and derived markers. Derived markers were divided into groups based on how
they
correlated to each of the markers in the top-performing (based on AUC) ratio.
This ratio
approach provides the best diagnostic power with respect to AUC for
separating: healthy and
post-surgical (PS) (also referred to herein as "inSIRS") conditions; healthy
and sepsis (also
referred to herein as "ipSIRS"); inSIRS and ipSIRS; mild ipSIRS and severe
ipSIRS; mild
ipSIRS and septic shock; and severe ipSIRS and septic shock.
Clinical Trials
[0538] Clinical trials were performed to determine whether certain mRNA
transcripts could
distinguish between healthy controls and various patient groups and within
patient groups.
Intensive care sepsis, post-surgical and inSIRS patients, as well as healthy
controls were

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 165 -
prospectively enrolled and attended a single visit where blood was collected
for gene
expression and mRNA analyses using Affymetrix exon arrays and/or quantitative
real-time
PCR (qRT-PCR). A definitive diagnosis of infection-positive SIRS (mild, severe
or shock)
or inSIRS was unlikely to be known at the time patients were enrolled, and
thus confirmation
of a diagnosis and the assignment of patients to the cohorts were made
retrospectively.
[0539] Patients who had clinical signs and/or symptoms of ipSIRS or inSIRS
were consented
and enrolled into the study as soon as possible after they had been
identified, in most cases
within 24 hours of admission. Final assessment of whether the participant had
inSIRS,
ipSIRS (mild, severe, or shock) was made retrospectively as clinical
information and blood
culture results became available.
[0540] Study participants were all over 18 years and either they or their
surrogate decision
maker signed and dated the clinical trial information sheet and consent form.
All of the
control participants were considered to be in good health based on a brief
physical
examination and their medical history at the time of enrolment.
[0541] Patients or their surrogate decision maker were offered the opportunity
to participate
in this study if the patient presented with signs and symptoms of either
inSIRS or ipSIRS at
the time of admission to ICU (using criteria based on the American College of
Physicians
and the Society of Critical Care Medicine standard definitions). That is,
inSIRS and ipSIRS
participants needed a variable combination of clinical conditions including
two or more of
the following within the last 24 hours: temperature >38 C or <36 C; heart rate
>90
beats/min; respiratory rate >20 breathes/min or a PaCO2 of <4.3kPa (<32 mm
Hg); and
evidence of a white blood cell count <4,000 cells/mm3 (<4 x 109 cells/L) or
>12,000
cells/mm3 (>12 x 109 cells/L) or >10% immature neutrophils (band forms).
Participants were
excluded if they had any chronic systemic immune-inflammatory disorders
including SLE,
Crohn's disease, insulin-dependent diabetes mellitus (IDDM); were transplant
recipients or
were currently receiving chemotherapy treatment for cancer. Most patients had
other
underlying co-morbidities. All study participants were 18 years of age or
older and had a
body mass index of less than 40.
[0542] Demography, vital signs measurements (blood pressure, heart rate,
respiratory rate,
oxygen saturation, temperature), hematology (full blood count), clinical
chemistry (urea,
electrolytes, liver function enzymes, blood glucose) as well as microbial
status was recorded.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 166 -
[0543] Blood was collected for the purpose of extraction of high quality RNA
into
PAXgeneTM tubes (PreAnalytix Inc., Valencia, CA, USA). Blood for bacterial
culture was
collected into BacTec Plus Aerobic (10m1) and BacTec Plus Anaerobic (10mL)
tubes
(Becton Dickinson) tubes for the detection of aerobic and anaerobic bacterial
growth
respectively.
[0544] A PAXgene blood RNA kit available from Qiagen Inc. (Valencia, CA, USA)
was
used to isolate total RNA from PAXgene tubes. Isolation begins with a
centrifugation step to
pellet nucleic acids in the PAXgene blood RNA tube. The pellet is washed and
re-suspended
and incubated in optimized buffers together with Proteinase K to bring about
protein
digestion. An additional centrifugation is carried out to remove residual cell
debris and the
supernatant is transferred to a fresh microcentrifuge tube. Ethanol is added
to adjust binding
conditions, and the lysate is applied to the PAXgene RNA spin column. During
brief
centrifugation, RNA is selectively bound to the silica-gel membrane as
contaminants pass
through. Remaining contaminants are removed in three efficient wash steps and
RNA is then
eluted in Buffer BR5. Determination of RNA quantity and quality was performed
using an
Agilent Bioanalyzer and Absorbance 260/280 ratio using a spectrophotometer.
Processing of Samples
[0545] Measurement of specific mRNA levels in a tissue sample can be achieved
using a
variety of technologies. A common and readily available technology that covers
most of the
known human mRNAs is GeneChip analysis using Affymetrix technology. Details
on the
technology and methodology can be found at www.affymetrix.com. GeneChip
analysis has
the advantage of being able to analyze thousands of RNA transcripts at a time.
Another
common and readily available technology is qPCR (quantitative polymerase chain
reaction),
which has the advantage of being able to analyze, in real-time and
quantitatively, hundreds of
RNA transcripts at a time. Details on one of these technologies (TaqMan ),
chemistries and
methodologies can be found on the Life Technologies website including
published protocols
entitled; "Protocol: Introduction to TaqMan SYBR Green Chemistries for Real-
Time PCR"
and "TaqMan Gene Expression Assays Protocol." Both GeneChip and qPCR were
used in
the discovery and proof-of-concept stages for biomarker identification. qPCR
was used
exclusively for biomarker feasibility testing.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 167 -
Analysis, Interpretation of Data and Selection of Biomarkers and Derived
Biomarkers
Healthy Control versus inSIRS
[0546] A list of 941 mRNA individual markers with an AUC of at least 0.7 for
separating the
two conditions of Healthy and inSIRS was generated. Figure 8A plots these
markers against
the AUC and Figure 8B is a box and whisker plot of the best mRNA biomarker for
separating
the two conditions (AGFG1 ¨ ArfGAP with FG repeats 1). The conditions of
Healthy and
inSIRS are perfectly separated when using this mRNA biomarker alone.
[0547] From the 941 individual markers at least 1000 derived markers (ratios)
were
generated that had an AUC of 1Ø A plot of the AUC of these derived markers
for separating
the conditions of Healthy and inSIRS is shown in Figure 8C with the top
performing ratio
shown as a box and whisker plot in Figure 8D. The AUC for AGFG1 and PVRIG
(poliovirus
receptor related immunoglobulin domain containing) has a ratio is 1Ø
[0548] All 941 individual markers were then broken into two groups ¨ those
that correlate to
AGFG1 and those that correlate to PVRIG. Figures 8E and 8F demonstrate the
correlation
between the two groups based on their similarity to either AGFG1 or PVRIG. In
these plots
the groups are referred to as either group 1 (AGFG1) or group 2 (PVRIG). It
can be seen that
each "group" contains those markers that are most highly correlated to each
other.
[0549] The markers in each "group" also correlate strongly (as demonstrated by
AUC greater
than 0.7 for all markers) to the condition being studied (in this instance
Healthy versus
inSIRS (PS)) as shown in Figures 8G and 8H.
[0550] By choosing mRNAs from these two groups to create derived markers a
better AUC
for separating Healthy and inSIRS is obtained than when markers are chosen
from within
groups (p< 2.558e-13) as demonstrated in Figure 81, which shows a greater
overall AUC is
obtained compared to when using markers from either group 1 or group 2 alone.
The mean
AUC for markers derived from groups 1 and 2 is over 0.97, whereas the mean AUC
for
markers derived from either group 1 or 2 alone is less than 0.9.
Healthy Control Versus ipSIRS
[0551] A list of 941 mRNA individual markers with an AUC of at least 0.7 for
separating the
two conditions of Healthy and ipSIRS was generated. Figure 9A plots these
markers against
the AUC and Figure 9B is a box and whisker plot of the best mRNA biomarker for
separating
the two conditions (LTBP3 ¨ latent transforming growth factor beta binding
protein 3). The

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 168 -
conditions of Healthy and ipSIRS are perfectly separated when using this mRNA
biomarker
alone.
[0552] From the 941 individual markers at least 1000 derived markers (ratios)
were
generated that had an AUC of 1Ø A plot of the AUC of these derived markers
for separating
the conditions of Healthy and inSIRS is shown in Figure 9C with the top
performing ratio
shown as a box and whisker plot in Figure 9D. The AUC for LTBP3 and LPHN1
(latrophilin
1) as a ratio is 1Ø
[0553] All 941 individual markers were then broken into two groups ¨ those
that correlate to
LTBP3 and those that correlate to LPHN1. Both plots in Figures 9E and 9F
demonstrate the
correlation between the two groups based on their similarity to either LTBP3
or LPHN1. It
can be seen that each "group" contains those markers that are most highly
correlated to each
other.
[0554] The markers in each "group" also correlate strongly (as demonstrated by
AUC greater
than 0.7 for all markers) to the condition being studied (in this instance
Healthy versus
ipSIRS (sepsis)), as shown in Figures 9G and 9H.
[0555] By choosing mRNAs from these two groups to create derived markers a
better AUC
for separating Healthy and ipSIRS is obtained than when markers are chosen
from within
groups (p< 2.2e-16) as demonstrated in Figure 91, which shows an improved AUC
compared
to using markers from either group 1 or group 2 alone. The mean AUC for
markers derived
from groups 1 and 2 is over 0.97, whereas the mean AUC for markers derived
from either
group 1 or 2 alone is less than 0.8.
inSIRS versus ipSIRS
[0556] A list of 359 mRNA individual markers with an AUC of at least 0.7 for
separating the
two conditions of inSIRS and ipSIRS was generated. Figure 10A plots these
markers against
the AUC and Figure 10B is a box and whisker plot of the best mRNA biomarker
for
separating the two conditions (PIWIL4 ¨ piwi-like RNA mediated gene silencing
4). The
conditions of inSIRS (PS) and ipSIRS (sepsis) are well separated when using
this mRNA
biomarker alone.
[0557] From the 359 individual markers 1000 derived markers (ratios) were
generated that
had an AUC greater than 0.9. A plot of the AUC of these derived markers for
separating the

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 169 -
conditions of inSIRS and ipSIRS is shown in Figure 10C with the top performing
ratio of
PLA2G7 (phospholipase A2, Group VII, (platelet activating factor acetyl
hydrolase, plasma))
and PLAC8 (placenta-specific 8) shown as a box and whisker plot in Figure 10D.
[0558] All 359 individual markers were then broken into two groups ¨ those
that correlate to
PLA2G7 and those that correlate to PLAC8. The plot in Figure 10E demonstrates
that the
markers in each "group" correlate strongly (as demonstrated by AUC greater
than 0.7 for all
markers) to the condition being studied (in this instance inSIRS (PS) versus
ipSIRS (sepsis)).
[0559] By choosing mRNAs from these two groups to create derived markers a
better AUC
for separating inSIRS (PS) and ipSIRS (sepsis) is obtained than when markers
are chosen
from within groups (p< 5.78e-5) as demonstrated in Figure 1OF compared to when
using
markers from either group 1 or group 2 alone. The mean AUC for markers derived
from
groups 1 and 2 is over 0.80, whereas the mean AUC for markers derived from
either group 1
or 2 alone is less than 0.8.
[0560] In an alternative embodiment, the markers were broken into four groups
¨ those that
correlate to CEACAM4 (bucket 3), those that correlate to LAMP] (bucket 4),
those that
correlate to PLA2G7 (bucket 1) and those that correlate to PLAC8 (bucket 2).
The plots in
Figure 10G demonstrate the markers in each group correlate strongly (as
demonstrated by
AUC greater than 0.7 for all markers) to the condition being studied (i.e.,
inSIRS (PS) versus
ipSIRS (sepsis)). Like the two-bucket embodiment discussed above, choosing
mRNAs from
four groups to create derived markers results in an overall better AUC for
separating inSIRS
(PS) and ipSIRS (sepsis), as compared to when markers are chosen from within
groups (p<
0.2564, as demonstrated in Figure 10H compared to when using markers from any
one of
groups 1 to 4. The mean AUC for markers derived from groups 1 to 4 is over 0.8
whereas
the mean AUC for markers derived from any one of groups 1 to 4 is less than
0.8
Mild ipSIRS versus Severe ipSIRS
[0561] A list of 66 mRNA individual markers with an AUC of at least 0.7 for
separating the
two conditions of mild ipSIRS and severe ipSIRS was generated. Figure 11A
plots these
markers against the AUC and Figure 11B is a box and whisker plot of the best
mRNA
biomarker for separating the two conditions (N4BP2L2 ¨ NEDD4 binding protein 2-
like 2).
The conditions of mild ipSIRS and severe ipSIRS are well separated when using
this mRNA
biomarker alone.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 170 -
[0562] From the 66 individual markers at least 1000 derived markers (ratios)
were generated
that had an AUC of 0.87. A plot of the AUC of these derived markers for
separating the
conditions of mild ipSIRS and severe ipSIRS is shown in Figure 11C with the
top performing
ratio shown as a box and whisker plot in Figure 11D. The AUC for N4BP2L2 and
ZC3H11A
(zinc finger CCCH-type containing 11A) as a ratio is 0.983.
[0563] All 66 individual markers were then broken into two groups ¨ those that
correlate to
N4BP2L2 and those that correlate to ZC3H11A. Both plots in Figures 11E and 11F

demonstrate the correlation between the two groups based on their similarity
to either
N4BP2L2 or ZC3H11A. In these plots the groups are referred to as either group
1
(N4BP2L2) or group 2 (ZC3H11A). It can be seen that each "group" contains
those markers
that are most highly correlated to each other. The markers in each "group"
also correlate
strongly (as demonstrated by AUC greater than 0.7 for all markers) to the
condition being
studied (in this instance mild ipSIRS versus severe ipSIRS).
[0564] By choosing mRNAs from these two groups to create derived markers a
better AUC
for separating mild ipSIRS and severe ipSIRS is obtained than when markers are
chosen from
within groups (p< 2.2e-16) as demonstrated in Figure 111, compared to when
using markers
from either group 1 or group 2 alone. The mean AUC for markers derived from
groups 1 and
2 is over 0.89, whereas the mean AUC for markers derived from either group 1
or 2 alone is
less than 0.6.
Mild ipSIRS versus ipSIRS ¨ shock
[0565] A list of 48 mRNA individual markers with an AUC of at least 0.7 for
separating the
two conditions of mild ipSIRS and ipSIRS - shock was generated. Figure 12A
plots these
markers against the AUC and Figure 12B is a box and whisker plot of the best
mRNA
biomarker for separating the two conditions (CD6 ¨ CD6 molecule). The
conditions of mild
ipSIRS and ipSIRS shock are well separated when using this mRNA biomarker
alone.
[0566] From the 48 individual markers at least 1000 derived markers (ratios)
were generated
that had an AUC of at least 0.793. A plot of the AUC of these derived markers
for separating
the conditions of mild ipSIRS and ipSIRS shock is shown in Figure 12C with the
top
performing ratio shown as a box and whisker plot in Figure 12D. The AUC for
VAMP2 and
UBAP1 (ubiquitin associated protein 1) as a ratio is 0.978.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 171 -
[0567] All 48 individual markers were then broken into two groups ¨ those that
correlate to
VAMP2 and those that correlate to UBAP1. Both plots in Figure 12E and 12F
demonstrate
the correlation between the two groups based on their similarity to either
VAMP2 or UBAP1.
In these plots the groups are referred to as either group 1 (VAMP2) or group 2
(UBAP1). It
can be seen that each "group" contains those markers that are most highly
correlated to each
other. The markers in each "group" also correlate strongly (as demonstrated by
AUC greater
than 0.7 for all markers) to the condition being studied (in this instance
mild ipSIRS versus
ipSIRS - shock).
[0568] By choosing mRNAs from these two groups to create derived markers a
better AUC
for separating mild ipSIRS and ipSIRS - shock is obtained than when markers
are chosen
from within groups (p< 2.2e-16) as demonstrated in Figure 121, compared to
when using
markers from either group 1 or group 2 alone. The mean AUC for markers derived
from
groups 1 and 2 is over 0.87, whereas the mean AUC for markers derived from
either group 1
or 2 alone is less than 0.65.
Severe ipSIRS versus ipSIRS ¨ shock
[0569] A list of 61 mRNA individual markers with an AUC of at least 0.7 for
separating the
two conditions of severe ipSIRS and ipSIRS - shock was generated. Figure 13A
plots these
markers against the AUC and Figure 13B is a box and whisker plot of the best
mRNA
biomarker for separating the two conditions (SIRPG ¨ signal regulatory protein
gamma). The
conditions of severe ipSIRS and ipSIRS - shock are well separated when using
this mRNA
biomarker alone.
[0570] From the 61 individual markers at least 1000 derived markers (ratios)
were generated
that had an AUC of at least 0.821. A plot of the AUC of these derived markers
for separating
the conditions of severe ipSIRS and ipSIRS - shock is shown in Figure 13C with
the top
performing ratio shown as a box and whisker plot in Figure 13D. The AUC for
GATA3
(GATA binding protein 3) and MECOM (MDS1 and EVI1 complex locus) as a ratio is
0.936.
[0571] All 61 individual markers were then broken into two groups ¨ those that
correlate to
GATA3 and those that correlate to MECOM. Both plots in Figures 13E and 13F
demonstrate
the correlation between the two groups based on their similarity to either
GATA3 or
MECOM. In these plots the groups are referred to as either group 1 (GATA3) or
group 2
(MECOM). It can be seen that each "group" contains those markers that are most
highly

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 172 -
correlated to each other. The markers in each "group" also correlate strongly
(as
demonstrated by AUC greater than 0.7 for all markers) to the condition being
studied (in this
instance severe ipSIRS versus ipSIRS - shock).
[0572] By choosing mRNAs from these two groups to create derived markers a
better AUC
for separating severe ipSIRS and ipSIRS - shock is obtained than when markers
are chosen
from within groups (p< 2.2e-16) as demonstrated in Figure 131 compared to when
using
markers from either group 1 or group 2 alone. The mean AUC for markers derived
from
groups 1 and 2 is over 0.82, whereas the mean AUC for markers derived from
either group 1
or 2 alone is less than 0.7.
Signature Usage
[0573] Use of the above described markers and resulting signatures in patient
populations
and benefits in respect of differentiating inSIRS and ipSIRS, will now be
described.
[0574] An assay capable of differentiating patients with inSIRS and ipSIRS can
be used in
multiple patient populations including:
1) Intensive Care Unit (medical and surgical ICU)
2) Post-surgical and medical wards
3) Emergency Department
4) Medical clinics.
[0575] Patients admitted to intensive care (ICU) often have ipSIRS, or develop
ipSIRS
during their ICU stay. The ultimate aim of intensive care is to ensure the
patient survives and
is discharged to a general ward in the minimum time. Patients in intensive
care with
diagnosed ipSIRS are usually administered a number of therapeutic compounds ¨
many of
which have opposing actions on the immune system and many of which could be
counterproductive depending on the severity of ipSIRS (mild sepsis, severe
sepsis, septic
shock). Monitoring intensive care patients on a regular basis with biomarkers
of the present
invention will allow medical practitioners to differentiate inSIRS from ipSIRS
and determine
the stage of ipSIRS and hence choice of therapies, when to start and stop
therapies, and
patient management procedures, and ultimately response to therapy. Information
provided by
these biomarkers will therefore allow medical intensivists to tailor and
modify therapies to
ensure patients survive and spend less time in intensive care. Less time in
intensive care leads

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 173 -
to considerable savings in medical expenses including through less occupancy
time and
appropriate use and timing of medications.
[0576] Surgical and general medical patients often develop inSIRS post-surgery
or as a
consequence of their condition or procedures and have a higher risk of
developing ipSIRS.
Post-operative and medical care in such patients therefore involves monitoring
for signs of
inSIRS and ipSIRS and differentiating between these two conditions. The
treatment and
management of inSIRS and ipSIRS patients post-surgically and in general wards
is different,
since inSIRS patients can be put on mild anti-inflammatory drugs or anti-
pyretics and ipSIRS
patients must be started on antibiotics as soon as possible for best outcomes.
Monitoring
post-surgical and medical patients on a regular basis with biomarkers of the
present invention
will allow nursing and medical practitioners to differentiate inSIRS and
ipSIRS at an early
stage and hence make informed decisions on choice of therapies and patient
management
procedures, and ultimately response to therapy. Information provided by these
biomarkers
will therefore allow medical practitioners to tailor and modify therapies to
ensure patients
recover quickly from surgery or other condition and do not develop ipSIRS.
Less time in
hospital and less complications leads to considerable savings in medical
expenses including
through less occupancy time and appropriate use and timing of medications.
[0577] Further, patients presenting to emergency departments often have a
fever, which is
one (of four) of the clinical signs of inSIRS. Such patients need to be
assessed to determine if
they have either inSIRS or ipSIRS. As mentioned above, the treatment and
management of
pyretic, inSIRS and septic patients are different. By way of example, a
patient with a fever
without other inSIRS clinical signs and no obvious source of infection may be
sent home, or
provided with other non-hospital services, without further hospital treatment.
However, a
patient with a fever may have early ipSIRS and not admitting such a patient
may put their life
at risk. Because these biomarkers can differentiate inSIRS and ipSIRS they
will allow
medical practitioners to triage emergency department patients quickly and
effectively.
Accurate triage decision-making insures that patients requiring hospital
treatment are given
it, and those that don't are provided with other appropriate services.
[0578] Further still, patients presenting to medical clinics often have any
one of the four
clinical signs of inSIRS (increased heart rate, increased respiratory rate,
abnormal white
blood cell count, fever or hypothermia). Many different clinical conditions
can present with

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 174 -
one of the four clinical signs of inSIRS and such patients need to be assessed
to determine if
they have either inSIRS or ipSIRS and to exclude other differential diagnoses.
By way of
example, a patient with colic might also present with clinical signs of
increased heart rate.
Differential diagnoses could be (but not limited to) appendicitis,
urolithiasis, cholecystitis,
pancreatitis, enterocolitis. In each of these conditions it would be important
to determine if
there was a systemic inflammatory response (inSIRS) or whether an infection
was
contributing to the systemic response to the condition (ipSIRS). The treatment
and
management of patients with and without systemic inflammation and/or infection
are
different. Because these biomarkers can differentiate patients with a systemic
inflammatory
response to infection from those with a systemic inflammatory response without
infection
(inSIRS and ipSIRS), and determine the degree of systemic involvement, the use
of them will
allow medical practitioners to determine the next medical procedure(s) to
perform to
satisfactorily resolve the patient issue.
Determining the Extent of Systemic Inflammation in Sick Patients and Those
With inSIRS and
ipSIRS
[0579] As mentioned above, patients presenting to medical clinics often have
any one of the
four clinical signs of inSIRS. However, many different clinical conditions can
present with
one of the four clinical signs of inSIRS and such patients need to be assessed
to determine if
they have inSIRS, and if so the extent of inSIRS, or ipSIRS, and if so the
extent of ipSIRS,
and to exclude other differential diagnoses.
[0580] By way of example, a patient with respiratory distress is likely to
present with clinical
signs of increased respiratory rate. Differential diagnoses could be (but not
limited to)
asthma, pneumonia, congestive heart failure, physical blockage of airways,
allergic reaction,
collapsed lung, pneumothorax. In each of these conditions it would be
important to determine
if there was a systemic inflammatory response (inSIRS) or whether an infection
was
contributing to the condition. The treatment and management of patients with
and without
systemic inflammation and/or infection are different. Because these biomarkers
can
differentiate patients with a systemic inflammatory response to infection from
those with a
systemic inflammatory response without infection (inSIRS and ipSIRS), and
determine the
degree of systemic involvement, the use of them will allow medical
practitioners to
determine the next medical procedure(s) to perform to satisfactorily resolve
the patient issue.
Patients with a collapsed lung, pneumothorax or a physical blockage are
unlikely to have a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 175 -
large systemic inflammatory response and patients with congestive heart
failure, allergic
reaction or asthma are unlikely to have a large systemic inflammatory response
due to
infection. The extent of both inSIRS and ipSIRS, as indicated by biomarkers
presented in
this patent, also provides clinicians with information on next treatment and
management
steps. For example, a patient with respiratory distress and a strong biomarker
response
indicating ipSIRS is likely to be immediately hospitalized, placed on
antibiotics and a chest
X-ray performed. A patient with respiratory distress and a strong biomarker
response
indicating inSIRS but not ipSIRS is likely to be hospitalized and chest X-
rayed along with
other investigative diagnostic procedures, such as Mill, ECG, and angiogram. A
patient with
respiratory distress with a short history and no inSIRS or ipSIRS is likely to
undergo further
examination at a local clinic rather than requiring hospitalization.
[0581] Again, and as mentioned above, patients presenting to emergency
departments often
have a fever, which is one (of four) of the clinical signs of inSIRS. Such
patients need to be
assessed to determine if they have either inSIRS or ipSIRS. Further it is
important to
determine how sick they are to be able to make a Judgment call on whether to
admit the
patient or not. Accurate triage decision-making insures that patients
requiring hospital
treatment are given it, and those that don't are provided with other
appropriate services.
[0582] Patients in ICU often have inSIRS and ipSIRS and it is important to
differentiate
these two conditions as treatment regimens differ. In patients with inSIRS it
is important to
determine the extent of the inflammatory response so that appropriate
treatments and
management regimens can be put in place. For example, a patient newly
determined to have
inSIRS that is not extensive may be able to be put on mild medication such a
non-steroidal
anti-inflammatory. A patient newly determined to have extensive inSIRS (e.g.
trauma) may
require stronger anti-inflammatory medication such as steroids to reduce the
potential impact
of the side effects of inflammation (swelling). In patients with ipSIRS it is
also important to
determine the extent of the inflammatory response to infection so that
appropriate treatments
and management regimens can be put in place or stopped. For example, for a
patient with a
persistent strong ipSIRS response the clinician may consider either changing,
or adding to,
the antibiotic treatment regimen in the absence of traditional bacterial
culture and sensitivity
results. Further, patients that are known to have had ipSIRS and have been on
antimicrobial
therapy for an extended period but have since demonstrated (by testing using
biomarkers)

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 176 -
that they no longer have either an inSIRS or ipSIRS can therefore be safely
taken off
intravenous antibiotics.
Determining the Severity of ipSIRS
[0583] Patients admitted to intensive care (ICU) often have ipSIRS, or develop
ipSIRS
during their ICU stay. It is known that the severity of sepsis can be
considered to be a
continuum from less severe, or sepsis, to more severe, or severe sepsis, to
the most severe, or
septic shock. More severe sepsis (ipSIRS) requires more aggressive, immediate
and tailored
intervention compared to sepsis (although all are acute conditions). Patients
in intensive care
with diagnosed ipSIRS are usually administered a number of therapeutic
compounds ¨ many
of which have opposing actions on the immune system and many of which could be

counterproductive depending on the severity of ipSIRS (sepsis, severe sepsis,
septic shock).
Monitoring intensive care patients on a regular basis with biomarkers of the
present invention
will allow medical practitioners to determine the severity of ipSIRS (mild,
severe or shock)
and hence choice of therapies and patient management procedures, and
ultimately response to
therapy. Information provided by these biomarkers disclosed herein will
therefore allow
medical practitioners to tailor, modify or cease therapies and/or care to
ensure patients
survive and spend less time in intensive care. Less time in intensive care
leads to
considerable savings in medical expenses including through less occupancy time
and
appropriate use and timing of medications.
First Example Workflow
[0584] A first example workflow will now be described. The workflow involves
up to seven
steps depending upon availability of automated platforms. The assay uses
quantitative, real-
time determination of the amount of each transcript in the sample based on the
detection of
fluorescence on a qRT-PCR instrument (e.g. Applied Biosystems 7500 Fast Dx
Real-Time
PCR Instrument, Applied Biosystems, Foster City, CA, catalogue number 440685;
K082562). Transcripts are each amplified, detected, and quantified in a
separate reaction well
using a probe that is visualized in the FAM channel (by example). The reported
score is
calculated using interpretive software provided separately to the kit but
designed to integrate
with RT-PCR machines.
[0585] The workflow below describes the use of manual processing and a pre-
prepared kit.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
-177 -
Pre-analytical
[0586] Blood collection
[0587] Total RNA isolation
Analytical
[0588] Reverse transcription (generation of cDNA)
[0589] qPCR preparation
[0590] qPCR
[0591] Software, Interpretation of Results and Quality Control
[0592] Output.
Kit Contents
[0593] Diluent
[0594] RT Buffer
[0595] RT Enzyme Mix
[0596] qPCR Buffer
[0597] Primer/Probe Mix
[0598] AmpliTaq Gold (or similar)
[0599] High Positive Control
[0600] Low Positive Control
[0601] Negative Control
Blood Collection
[0602] The specimen used is a 2.5mL sample of blood collected by venipuncture
using the
PAXgene@ collection tubes within the PAXgene@ Blood RNA System (Qiagen, kit
catalogue # 762164; Becton Dickinson, Collection Tubes catalogue number
762165;
K042613). An alternate collection tube is Tempus@ (Life Technologies).
Total RNA Isolation
[0603] Blood (2.5mL) collected into a PAXgene RNA tube is processed according
to the
manufacturer's instructions. Briefly, 2.5mL sample of blood collected by
venipuncture using
the PAXgeneTm collection tubes within the PAXgeneTm Blood RNA System (Qiagen,
kit
catalogue # 762164; Becton Dickinson, Collection Tubes catalogue number
762165;
K042613). Total RNA isolation is performed using the procedures specified in
the
PAXgeneTm Blood RNA kit (a component of the PAXgeneTm Blood RNA System). The

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 178 -
extracted RNA is then tested for purity and yield (for example by running an A
260/280 ratio
using a Nanodrop (Thermo Scientific)) for which a minimum quality must be
(ratio > 1.6).
RNA should be adjusted in concentration to allow for a constant input volume
to the reverse
transcription reaction (below). RNA should be processed immediately or stored
in single-use
volumes at or below -70 C for later processing.
Reverse Transcription
[0604] Determine the appropriate number of reaction equivalents to be prepared
(master mix
formulation) based on a plate map and the information provided directly below.
Each clinical
specimen is run in singleton.
a) Each batch run must include the following specimens:
b) High Control, Low Control, Negative Control, and No Template Control (Test
Diluent instead of sample) in singleton each
Program the ABI 7500 Fast Dx Instrument as detailed below.
c) Launch the software.
d) Click Create New Document
e) In the New Document Wizard, select the following options:
i) Assay: Standard Curve (Absolute Quantitation)
ii) Container: 96-Well Clear
iii) Template: Blank Document (or select a laboratory-defined template)
iv) Run Mode: Standard 7500
v) Operator: Enter operator's initials
vi) Plate name: [default]
f) Click Finish
g) Select the Instrument tab in the upper left
h) In the Thermal Cycler Protocol area, Thermal Profile tab, enter the
following
times:
i) 25 C for 10 minutes
ii) 45 C for 45 minutes
iii) 93 C for 10 minutes
iv) Hold at 25 C for 60 minutes

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 179 -
[0605] In a template-free area, remove the test Diluent and RT-qPCR Test RT
Buffer to
room temperature to thaw. Leave the RT-qPCR Test RT Enzyme mix in the freezer
and/or on
a cold block.
[0606] In a template-free area, assemble the master mix in the order listed
below.
RT Master Mix ¨ Calculation:
Per well x N
RT-qPCR Test RT Buffer 3.5 tL 3.5 x N
RT-qPCR Test RT Enzyme mix 1.5 1.5 x N
Total Volume 5 u1_, 5 x N
[0607] Gently vortex the master mix then pulse spin. Add the appropriate
volume (5 ilL) of
the RT Master Mix into each well at room temperature.
[0608] Remove clinical specimens and control RNAs to thaw. (If the specimens
routinely
take longer to thaw, this step may be moved upstream in the validated method.)
[0609] Vortex the clinical specimens and control RNAs, then pulse spin. Add 10
!IL of
control RNA or RT-qPCR Test Diluent to each respective control or negative
well.
[0610] Add 10 !IL of sample RNA to each respective sample well (150 ng total
input for RT;
0D260/0D280 ratio greater than 1.6). Add 10 !IL of RT-qPCR Test Diluent to the
respective
NTC well.
[0611] Note: The final reaction volume per well is 15 L.
Samples
RT Master Mix 5 !IL
RNA sample 10 !IL
Total Volume (per well) 15 u1_,
[0612] Mix by gentle pipetting. Avoid forming bubbles in the wells.
[0613] Cover wells with a seal.
[0614] Spin the plate to remove any bubbles (1 minute at 400 x g).
[0615] Rapidly transfer to ABI 7500 Fast Dx Instrument pre-programmed as
detailed above.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 180 -
[0616] Click Start. Click Save and Continue. Before leaving the instrument, it
is
recommended to verify that the run started successfully by displaying a time
under Estimated
Time Remaining.
[0617] qPCR master mix may be prepared to coincide roughly with the end of the
RT
reaction. For example, start about 15 minutes before this time. See below.
[0618] When RT is complete (i.e. resting at 25 C; stop the hold at any time
before 60
minutes is complete), spin the plate to collect condensation (1 minute at 400
x g).
qPCR Preparation
[0619] Determine the appropriate number of reaction equivalents to be prepared
(master mix
formulation) based on a plate map and the information provided in RT
Preparation above.
[0620] Program the ABI 7500 Fast Dx with the settings below.
a) Launch the software.
b) Click Create New Document
c) In the New Document Wizard, select the following options:
i) Assay: Standard Curve (Absolute Quantitation)
ii) Container: 96-Well Clear
iii) Template: Blank Document (or select a laboratory-defined template)
iv) Run Mode: Standard 7500
v) Operator: Enter operator's initials
vi) Plate name: Enter desired file name
d) Click Next
e) In the Select Detectors dialog box:
i) Select the detector for the first biomarker, and then click Add>>.
ii) Select the detector second biomarker, and then click Add> , etc.
iii) Passive Reference: ROX
f) Click Next
g) Assign detectors to appropriate wells according to plate map.
i) Highlight wells in which the first biomarker assay will be assigned
ii) Click use for the first biomarker detector
iii) Repeat the previous two steps for the other biomarkers
iv) Click Finish

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 181 -
h) Ensure that the Setup and Plate tabs are selected
i) Select the Instrument tab in the upper left
j) In the Thermal Cycler Protocol area, Thermal Profile tab, perform the
following
actions, with the results shown in Figure 14:
i) Delete Stage 1 (unless this was completed in a laboratory-defined
template).
ii) Enter sample volume of 25 p.L.
iii) 95 C 10 minutes
iv) 40 cycles of 95 C for 15 seconds, 63 C for 1 minute
v) Run Mode: Standard 7500
vi) Collect data using the "stage 2, step 2 (63.0@1:00)" setting
k) Label the wells as below using this process: Right click over the plate
map, then
select Well Inspector. With the Well Inspector open, select a well or wells.
Click
back into the Well Inspector and enter the Sample Name. Close the Well
Inspector
when completed.
i) CONH for High Control
ii) CONL for Low Control
iii) CONN for Negative Control
iv) NTC for No Template Control
v) [Accession ID] for clinical specimens
1) Ensure that detectors and quenchers are selected as listed below.
i) FAM for CEACAM biomarker 1; quencher=none
ii) FAM for LAMP1 biomarker 2; quencher=none, etc.
iii) FAM for PLA2G7; quencher=none
iv) FAM for PLAC8; quencher=none
v) Select "ROX" for passive reference
qPCR
[0621] In a template-free area, remove the assay qPCR Buffer and assay
Primer/Probe Mixes
for each target to room temperature to thaw. Leave the assay AmpliTaq Gold in
the freezer
and/or on a cold block.
[0622] Still in a template-free area, prepare qPCR Master Mixes for each
target in the listed
order at room temperature.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 182 -
qPCR Master Mixes ¨ Calculation Per Sample
Per well x N
qPCR Buffer 11 !IL 11 xN
Primer/Probe Mix 3.4 tL 3.4 x N
AmpliTaq Gold 0.6 tL 0.6 x N
Total Volume 15 pt 15 x N
[0623] Gently mix the master mixes by flicking or by vortexing, and then pulse
spin. Add 15
!IL of qPCR Master Mix to each well at room temperature.
[0624] In a template area, add 130 !IL of SeptiCyte Lab Test Diluent to each
cDNA product
from the RT Reaction. Reseal the plate tightly and vortex the plate to mix
thoroughly.
[0625] Add 10 !IL of diluted cDNA product to each well according to the plate
layout.
[0626] Mix by gentle pipetting. Avoid forming bubbles in the wells.
[0627] Cover wells with an optical seal.
[0628] Spin the plate to remove any bubbles (1 minute at 400 x g).
[0629] Place on real-time thermal cycler pre-programmed with the settings
above.
[0630] Click Start. Click Save and Continue. Before leaving the instrument, it
is
recommended to verify that the run started successfully by displaying a time
under Estimated
Time Remaining.
[0631] Note: Do not open the qPCR plate at any point after amplification has
begun. When
amplification has completed, discard the unopened plate.
Software, Interpretation of Results and Quality Control
[0632] Software is specifically designed to integrate with the output of PCR
machines and to
apply an algorithm based on the use of multiple biomarkers. The software takes
into account
appropriate controls and reports results in a desired format.
[0633] When the run has completed on the ABI 7500 Fast Dx Instrument, complete
the steps
below in the application 7500 Fast System with 21 CFR Part 11 Software, ABI
software
SDS v1.4.
[0634] Click on the Results tab in the upper left corner.
[0635] Click on the Amplification Plot tab in the upper left corner.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 183 -
[0636] In the Analysis Settings area, select an auto baseline and manual
threshold for all
targets. Enter 0.01 as the threshold.
[0637] Click on the Analyze button on the right in the Analysis Settings area.
[0638] From the menu bar in the upper left, select File then Close.
[0639] Complete the form in the dialog box that requests a reason for the
change. Click OK.
[0640] Transfer the data file (.sds) to a separate computer running the
specific assay RT-
qPCR Test Software.
[0641] Launch the assay RT-qPCR Test Software. Log in.
[0642] From the menu bar in the upper left, select File then Open.
[0643] Browse to the location of the transferred data file (.sds). Click OK.
[0644] The data file will then be analyzed using the assay's software
application for
interpretation of results.
Interpretation of Results and Quality Control
Results
[0645] Launch the interpretation software. Software application instructions
are provided
separately.
[0646] Following upload of the .sds file, the Software will automatically
generate classifier
scores for controls and clinical specimens.
[0647] Controls
[0648] The Software compares each CON (control) specimen (CONH, CONL, CONN) to
its
expected result. The controls are run in singleton.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 184 -
Control specimen
Designation Name Expected result
CONH High Control Score range
CONL Low Control Score range
CONN Negative Control Score range
NTC No Template Fail (no Ct for all
Control targets)
[0649] If CONH, CONL, and/or CONN fail the batch run is invalid and no data
will be
reported for the clinical specimens. This determination is made automatically
by the
interpretive software. The batch run should be repeated starting with either a
new RNA
preparation or starting at the RT reaction step.
[0650] If NTC yields a result other than Fail (no Ct for all targets), the
batch run is invalid
and no data may be reported for the clinical specimens. This determination is
made by visual
inspection of the run data. The batch run should be repeated starting with
either a new RNA
preparation or starting at the RT reaction step.
[0651] If a second batch run fails, please contact technical services. If both
the calibrations
and all controls are valid, then the batch run is valid and specimen results
will be reported.
[0652] Specimens
[0653] Note that a valid batch run may contain both valid and invalid specimen
results.
[0654] Analytical criteria (e.g. Ct values) that qualify each specimen as
passing or failing
(using pre-determined data) are called automatically by the software.
[0655] Scores out of range ¨ reported.
[0656] Quality Control
[0657] Singletons each of the Negative Control, Low Positive Control, and High
Positive
Control must be included in each batch run. The batch is valid if no flags
appear for any of
these controls.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 185 -
[0658] A singleton of the No Template Control is included in each batch run
and Fail (no Ct
for all targets) is a valid result indicating no amplifiable material was
detectable in the well.
[0659] The negative control must yield a Negative result. If the negative
control is flagged
as Invalid, then the entire batch run is invalid.
[0660] The low positive and high positive controls must fall within the
assigned ranges. If
one or both of the positive controls are flagged as Invalid, then the entire
batch run is invalid.
Example Output
[0661] A possible example output from the software is presented below in
Figure 15. The
format of such a report depends on many factors including; quality control,
regulatory
authorities, cut-off values, the algorithm used, laboratory and clinician
requirements,
likelihood of misinterpretation.
[0662] In this instance the assay is called "SeptiCyte Lab Test". The result
is reported as a
number (5.8), a call ("Sepsis Positive"), a position on a 0-12 scale, and a
probability of the
patient having sepsis based on historical results and the use of a pre-
determined cut-off (using
results from clinical trials). Results of controls within the assay are also
reported. Other
information that could be reported might include: previous results and date
and time of such
results, probability of severe sepsis or septic shock, a scale that provides
cut-off values for
historical testing results that separate the conditions of healthy, inSIRS and
ipSIRS (mild,
severe and shock) such that those patients with higher scores are considered
to have more
severe inSIRS or ipSIRS.
Second Example Workflow
[0663] A second example workflow will now be described. Machines have been,
and are
being, developed that are capable of processing a patient sample at point-of-
care, or near
point-of-care. Such machines require few molecular biology skills to run and
are aimed at
non-technical users. The idea is that the sample would be pipetted directly
into a disposable
cartridge that is then inserted into the machine. The user presses "Start" and
within 2-3 hours
a result is generated. The cartridge contains all of the required reagents to
perform Steps 2-5
in the example workflow above and the machine has appropriate software
incorporated to
allow Steps 6 and 7 to be performed.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 186 -
[0664] Fresh, whole, anti-coagulated blood can be pipetted into an Idylla
Cartridge (Biocartis
NV) or similar (Unyvero, Curetis AG; Enigma ML, Enigma Diagnostics; DiagCore,
STAT
Diagnostica; Savannah, Quidel Corp; ePlex, GenMark Dx), and on-screen
instructions on the
Idylla machine followed to test for differentiating inSIRS and ipSIRS (by
example). Inside
the Idylla machine RNA is first extracted from the whole blood and is then
converted into
cDNA. The cDNA is then used in qRT-PCR reactions. The reactions are followed
in real
time and Ct values calculated. On-board software generates a result output
(see Figure XX).
Appropriate quality control measures for RNA quality, no template controls,
high and low
template controls and expected Ct ranges ensure that results are not reported
erroneously.
Example Biomarker Ratios
[0665] Example biomarker ratios (the top 12 based on AUC) that are capable of
separating
different conditions are shown in the box and whisker plots as listed below,
with each
showing perfect separation.
= Figures 16A to 16L show Healthy Versus inSIRS (Post-Surgical)
= Figures 17A to 17L show Healthy Versus ipSIRS (Sepsis)
= Figures 18A to 18L show inSIRS (Post-Surgical) Versus ipSIRS (Sepsis)
= Figures 19A to 19L show Sepsis Versus Severe Sepsis
= Figures 20A to 20L show Severe Sepsis Versus Septic Shock
= Figures 21A to 21L show Sepsis Versus Septic Shock
Example Algorithm Combining Biomarker Ratios
[0666] Biomarker ratios (derived markers) can be used in combination to
increase the
diagnostic power for separating various conditions. Determining which markers
to use, and
how many, for separating various conditions can be achieved by calculating
Area Under
Curve (AUC).
[0667] Figure 22 shows the effect on AUC (in this instance for separating
inSIRS and
ipSIRS) of adding biomarkers to the diagnostic signature. Diagnostic power
significantly
increases (adjusted p-value = 0.0175) between a single mRNA biomarker (in this
instance
PLA2G7, AUC of 0.88, 95% CI 0.79 - 0.97) compared to the power of the two best

performing markers in combination (in this instance PLA2G7 and PLAC8, AUC of
0.96, 95%
CI 0.91 - 1.0). Combinations of two, three, four and five biomarkers produced
equally as

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 187 -
good differentiation of inSIRS and ipSIRS without significant differences. For
commercial
development of derived markers other factors come into play such as cost-
effectiveness,
assay complexity and capabilities of the qRT-PCR platform.
[0668] In this example, the addition of markers beyond 3 or 4 does not
significantly improve
performance and, conversely, a decline in AUC is observed in signatures of >5
genes
probably because when a statistical model is forced to include biomarkers that
add little
additional information data over-fitting and addition of noise occurs.
[0669] As such, and by example, a 4-gene signature (0.986, 95% CI 0.964-1.00)
offers the
appropriate balance between simplicity, practicality and commercial risk for
separating
inSIRS and ipSIRS. Further, an equation using four markers weighs each
biomarker equally
which also provides additional robustness in cases of analytical or clinical
variability.
[0670] One example equation that provides good diagnostic power for separating
inSIRS and
ipSIRS (amongst others) is:
Diagnostic Score = (PLA2G7 ¨ PLAC8)+ (CEACAM4 ¨ LAMP])
[0671] The value for each biomarker is a Ct value from a PCR. When clinical
samples from
patients with inSIRS and ipSIRS were tested using these four markers in a PCR
the Ct values
for each of the markers was found to fall between 26 and 34. In this patient
population the
first biomarker within each bracket pair has a higher value than the second
biomarker within
each bracket pair. Thus, the "Diagnostic Score" has been found to have values
between 0
and 12. However, in theory the "Diagnostic Score could potentially be highest
Ct value +/-
highest Ct value.
[0672] In Figure 23 shows results of PCR and the use of the above algorithm
have been
calculated for two patient populations (N=63 for "Discovery" and N=70 for
"Feasibility").
Each patient was clinically and retrospectively (note, not at the time the
sample was taken)
confirmed as having either inSIRS (black dots) or ipSIRS (red dots). Each
patient sample has
also had a SeptiCyte score calculated (Y axis on left hand side). On a scale
of 0-12 it can be
seen that patients with confirmed ipSIRS (red dots) obtain a higher Diagnostic
Score
compared to those with confirmed inSIRS. Further, it can be seen that an
arbitrary cut-off
line can be drawn that more or less separates the two conditions depending
upon the desired
false negative or false positive rate (compared to a retrospective diagnosis
of inSIRS or

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 188 -
ipSIRS using clinical data). In this instance the line is drawn at a
"SeptiCyte Score" of 4
such that the number of false negative ipSIRS calls in the Discovery Dataset
is zero and the
number of false negative ipSIRS calls in the Feasibility Dataset is 2.
Conversely the number
of false positive ipSIRS calls in the Discovery Dataset is four and the number
of false
positive ipSIRS calls in the Feasibility Dataset is 9. Clearly in this
instance whether a patient
sample is false positive or false negative depends on the artificial gold
standard of a
retrospective clinical call of inSIRS or ipSIRS.
[0673] Accordingly, in one example, when used for determining a likelihood of
the subject
having inSIRS or ipSIRS, the method can include determining a first pair of
biomarker
values indicative of a concentration of polynucleotide expression products of
the PLA2G7
gene and PLAC8 gene, determining a second pair of biomarker values indicative
of a
concentration of polynucleotide expression products of the CEACAM4 gene and
LAMP]
gene and then determining an indicator using the first and second pairs of
biomarker values.
[0674] As previously discussed, the indicator could then be compared to
indicator references
specifically established to distinguish between inSIRS and ipSIRS.
[0675] An example process of a process for establishing indicator references
will now be
described in more details with reference to Figure 24.
[0676] In this example, at step 2400 the processing system 201 determines
reference data in
the form of measured biomarker values obtained for a reference population. The
reference
data may be acquired in any appropriate manner but typically this involves
obtaining gene
expression product data from a plurality of individuals.
[0677] In order to achieve this, gene expression product data are collected,
for example by
obtaining a biological sample, such as a peripheral blood sample, and then
performing a
quantification process, such as a nucleic acid amplification process,
including PCR
(Polymerase Chain Reaction) or the like, in order to assess the activity, and
in particular,
level or abundance of a number of reference biomarkers. Quantified values
indicative of the
relative activity are then stored as part of the reference data.
[0678] In one example, the measurements are received as raw data, which then
undergoes
preliminary processing. Such raw data corresponds to information that has come
from a

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 189 -
source without modification, such as outputs from instruments such as PCR
machines, array
(e.g., microarray) scanners, sequencing machines, clinical notes or any other
biochemical,
biological, observational data, or the like. This step can be used to convert
the raw data into
a format that is better suited to analysis. In one example this is performed
in order to
normalize the raw data and thereby assist in ensuring the biomarker values
demonstrate
consistency even when measured using different techniques, different
equipment, or the like.
Thus, the goal of normalization is to remove the variation within the samples
that is not
directly attributable to the specific analysis under consideration. For
example, to remove
variances caused by differences in sample processing at different sites.
Classic examples of
normalization include z-score transformation for generic data, or popular
domain specific
normalizations, such as RMA normalization for microarrays.
[0679] However, it will also be appreciated that in some applications, such as
a single sample
experiment run on a single data acquisition machine, this step may not
strictly be necessary,
in which case the function can be a Null function producing an output
identical to the input.
[0680] In one example, the preferred approach is a paired function approach
over log
normalized data. Log normalization is a standard data transformation on
microarray data,
because the data follow a log-normal distribution when coming off the machine.
Applying a
log transform turns the data into process-friendly normal data.
[0681] The individuals are selected to include individuals diagnosed with one
or more
conditions of interest, as well as healthy individuals The conditions are
typically medical,
veterinary or other health status conditions and may include any illness,
disease, stages of
disease, disease subtypes, severities of disease, diseases of varying
prognoses or the like, and
in the current example would include at least some individuals with inSIRS and
some
individuals with ipSIRS. In this regard, the individuals also typically
undergo a clinical
assessment allowing the conditions to be clinically identified, and with an
indication of any
assessment or condition forming part of the reference data.
[0682] The biomarker values measured will depend on the predominant condition
that is
being assessed so, for example, in the case of determining the likelihood of a
subject having
inSIRS or ipSIRS, the biomarkers used will be LAMP], CEACAM4, PLAC8 and
PLA2G7, as
discussed above.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 190 -
[0683] Once collected, the reference data can be stored in the database 211
allowing this to
be subsequently retrieved by the processing system 201 for subsequent
analysis, or could be
provided directly to the processing system 201 for analysis.
[0684] As part of the above process, at step 2410 the measurements are
validated using
traditional prior art techniques, to ensure that the measurements have been
performed
successfully, and hence are valid.
[0685] At step 2420, each individual with the reference population is
typically allocated to a
group. The groups may be defined in any appropriate manner and may be defined
based on
any one or more of an indication of a presence, absence, degree, stage,
severity, prognosis or
progression of a condition, other tests or assays, or measured biomarkers
associated with the
individuals.
[0686] For example, a first selection of groups may be to identify one or more
groups of
individuals suffering from SIRS, one or more groups of individuals suffering
ipSIRS, and
one or more groups of individuals suffering inSIRS. Further groups may also be
defined for
individuals suffering from other conditions. The groups may include
overlapping groups, so
for example it may be desirable to define groups of healthy individuals and
individuals
having SIRS, with further being defined to distinguish inSIRS patients from
ipSIRS patients,
as well as different degree of inSIRS or ipSIRS, with these groups having SIRS
in common,
but each group of patients differing in whether a clinician has determined the
presence of an
infection or not. Additionally, further subdivision may be performed based on
characteristics
of the individuals, phenotypic traits, measurement protocols or the like, so
groups could be
defined based on these parameters so that a plurality of groups of individuals
suffering from a
condition are defined, with each group relating to a different phenotypic
trait, measurement
protocol or the like.
[0687] It will also be appreciated, however, that identification of different
groups can be
performed in other manners, for example on the basis of particular activities
of biomarkers
within the biological samples of the reference individuals, and accordingly,
reference to
conditions is not intended to be limiting and other information may be used as
required.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 191 -
[0688] The manner in which classification into groups is performed may vary
depending on
the preferred implementation. In one example, this can be performed
automatically by the
processing system 201, for example, using unsupervised methods such as
Principal
Components Analysis (PCA), or supervised methods such as k-means or Self
Organizing
Map (SOM). Alternatively, this may be performed manually by an operator by
allowing the
operator to review reference data presented on a Graphical User Interface
(GUI), and define
respective groups using appropriate input commands.
[0689] At step 2430, first and second derived biomarker values are determined
representing
respective indicator values. The first and second indicator values In1, In2
are determined on a
basis of ratios of concentrations of first and second, and third and fourth
biomarkers
respectively:
= (PLA2G7I PLAC8)
1112 = (CEACAM41 LAMP])
[0690] The indicator values are then used to establish indicator references at
step 2440,
which are then used in analyzing measured indicator values for a subject to
establish a
likelihood of the subject having a condition.
[0691] In particular, indicator values for each reference group are
statistically analyzed to
establish a range or distribution of indicator values that is indicative of
each group, and an
example distribution is shown in Figure 26, as will be discussed in more
detail below.
[0692] A further example will now be described with reference to Figures 25A
and 25B.
[0693] In this example, at step 2500 a sample is acquired from the subject.
The sample could
be any suitable sample such as a peripheral blood sample, or the like,
depending on the nature
of the biomarker values being determined. At step 2505 the sample undergoes
preparation
allowing this to be provided to a measuring device and used in a
quantification process at
step 2510. For this purpose of this example, the quantification process
involves PCR
amplification, with the measuring device being a PCR machine, although other
suitable
techniques could be used. In this instance, amplifications times At(PLA2G7),
At(PLAC8),
At(CEACAM4), At(LAMP1) are determined for each of the four biomarkers at step
2515, with
the amplification times being transferred from the measuring device to the
processing system

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 192 -
201 allowing the processing system 201 to perform analysis of the
corresponding biomarker
values.
[0694] Accordingly, at step 2520 the processing system 201 calculates ratios
using the
amplifications times. In this regard, as the amplification times represent a
log value, the ratios
are determined by subtracting amplifications times as will be appreciated by a
person skilled
in the art.
[0695] Accordingly, in this example the indicator values would be determined
as follows:
At(PLA2G7) - At(PLAC8)
I112= At(CEACAM4) - At(LAMP1)
[0696] At step 2525 the processing system 201 determines an indicator value by
combining
the ratios for the indicator values, as follows:
In= Ini+ In2
[0697] The processing system 201 then compares the indicator value to one or
more
respective indicator references at step 2530.
[0698] As previously described, the indicator references are derived for a
reference
population and are used to indicate the likelihood of a subject suffering from
inSIRS or
ipSIRS. To achieve this, the reference population is grouped based on a
clinical assessment
into groups having / not having the conditions or a measure of severity, risk
or progression
stage of the condition, with this then being used to assess threshold
indicator values that can
distinguish between the groups or provide a measure of severity, risk or
progression stage.
[0699] The comparison is performed by comparing the indicator to an indicator
distribution
determined for each group in the reference population. In the current example,
there are two
reference groups, with one corresponding to individuals diagnosed with inSIRS
and the other
for individuals diagnosed with ipSIRS. In this instance, the results of the
comparison can be
used to determine a likelihood of the individual having ipSIRS as opposed to
inSIRS. This
can be achieved using a number of different methods, depending on the
preferred
implementation.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 193 -
[0700] An example of a reference distribution is shown in Figure 26, which
shows the
distribution of indicator values for a reference population containing both
inSIRS and ipSIRS
samples. The density (y axis) describes how common scores are in the reference
population.
In Figure 26, the most common values for the inSIRS population are in the
range 1 to 8, and
for the ipSIRS population are mostly in the range 5 to 13. By way of example,
let us assume
that the calculated indicator value for a new sample is 4. A value of 4 in the
inSIRS
population has a high density at this value (A), while the ipSIRS population
has a low density
at this value (B), meaning that this sample is more likely to be inSIRS.
Conversely, if a
sample has an indicator score of 10, this value in the ipSIRS reference
population has a high
density (C), while the inSIRS population has a low density (D), meaning this
it is more
probable that this sample with an indicator value of 10 belongs to the ipSIRS
population.
[0701] In practice this process can be performed by determining a basic
probability based on
score bands. For a given score band (i.e. 4-6), the proportion of individuals
with SIRS or
SEPSIS is calculated. For example, if 40% of the scores between 4 and 6 were
SEPSIS, then
if an subject has an indicator value between 4 and 6, they have a 40%
probability of
sepsis. Thus, for a given range within the reference distribution, the
probability of belonging
to one group or another (SIRS/SEPSIS) can simply be the proportion of that
group within the
range.
[0702] An alternative technique is a standard Bayes method. In this case, the
technique uses
a distribution of inSIRS scores, a distribution of ipSIRS scores and an
indicator value for the
subject. In this example, a standard score or equivalent is used to generate a
probability of
the indicator value belonging to the inSIRS distribution: pr(inSIRS) and
separately to the
ipSIRS distribution: pr(ipSIRS). The Bayes method is used to generate the
probability of
ipSIRS given the individual distributions.
[0703] Thus, given derived biomarker distributions for two or more groups
(i.e.
inSIRS/ipSIRS), the probability of membership for a single unknown sample into
each
distribution can be calculated (p-value) using for example a standard score (z-
score). Then
the p-values for each distribution can be combined into an overall probability
for each class
(i.e. inSIRS/ipSIRS) using for example Bayes rule or any other probability
calculation
method (including frequentist or empirical or machine learned methods).

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 194 -
[0704] Thus, once the indicator value has been derived and compared to the
indicator
distributions, the results of this comparison are used by the processing
system 201 to
calculate a likelihood of the subject having ipSIRS at step 2535, with this
being used to
generate a representation of the results at step 2540, which is provided for
display at step
2545, for example to a clinician or medical practitioner. This can be achieved
by displaying
the representation on a client device, such as part of an email, dashboard
indication or the
like.
[0705] An example of the representation is shown in Figures 27A and 27B.
[0706] In this example, the representation 2700 includes a pointer 2710 that
moves relative to
a linear scale 2720. The linear scale is divided into regions 2721, 2722,
2723, 2724 which
indicate whether the subject is suffering from level 1, 2, 3 or 4.
Corresponding indicator
number values are displayed at step 2730 with an indication of whether the
corresponding
value represents a likelihood of SIRS (inSIRS) or SEPSIS (ipSIRS) being shown
at step
2740. An alphanumeric indication of the score is shown at step 2751 together
with an
associated probability of the biological subject having SEPSIS at step 2752.
[0707] As shown in this example, regions of the linear scale where the pointer
is situated are
highlighted with the diagnosis that is most unlikely being greyed out to make
it absolutely
clear where the subject sits on the scale. This results in a representation
which when
displayed at step 2545 is easy for a clinician to readily understand and to
make a rapid
diagnosis.
[0708] It will be appreciated from the above that a method can be provided for
use in
assessing the likelihood of a biological subject having inSIRS or ipSIRS the
method
including, in one or more processing devices:
a) determining a pair of biomarker values, the pair of biomarker values being
selected from the group consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the PLA2G7 gene and PLAC8 gene;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide expression products of the CEACAM4 gene and LAMP] gene;

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 195 -
b) determining an indicator indicative of a ratio of the concentrations of the
polynucleotide expression products using the pair of biomarker values;
c) retrieving previously determined first and second indicator references from
a
database, the first and second indicator references being determined based on
indicators determined from first and second groups of a reference population,
one
of the groups consisting of individuals diagnosed with the medical condition;
d) comparing the indicator to the first and second indicator references;
e) using the results of the comparison to determine a probability indicative
of the
subject having the medical condition; and,
f) generating a representation of the probability, the representation being
displayed
to a user to allow the user to assess the likelihood of a biological subject
having at
least one medical condition.
[0709] Similarly apparatus can be provided for determining the likelihood of a
biological
subject having inSIRS or ipSIRS, the apparatus including:
a) a sampling device that obtains a sample taken from a biological subject,
the
sample including polynucleotide expression products;
b) a measuring device that quantifies polynucleotide expression products
within the
sample to determine a pair of biomarker values, the pair of biomarker values
being selected from the group consisting of:
i) a first pair of biomarker values indicative of a concentration of
polynucleotide
expression products of the PLA2G7 gene and PLAC8 gene;
ii) a second pair of biomarker values indicative of a concentration of
polynucleotide expression products of the CEACAM4 gene and LAMP] gene;
c) at least one processing device that:
i) receives an indication of the pair of biomarker values from the measuring
device;
ii) determines an indicator using a ratio of the concentration of the first
and
second polynucleotide expression products using the biomarker values; and,
iii) compares the indicator to at least one indicator reference; and,
iv) determines a likelihood of the subject having the at least one medical
condition using the results of the comparison; and,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 196 -
v) generates a representation of the indicator and the likelihood for display
to a
user.
[0710] A further method that can be provided includes differentiating between
inSIRS and
ipSIRS in a biological subject, the method including:
a) obtaining a sample taken from a biological subject showing a clinical sign
of
SIRS, the sample including polynucleotide expression products;
b) in a measuring device:
i) amplifying at least some polynucleotide expression products in the
sample;
ii) determining an amplification amount representing a degree of amplification

required to obtain a defined level of polynucleotide expression products
including:
(1) amplification amounts for a first pair of polynucleotide expression
products of the PLA2G7 gene and PLAC8 gene;
(2) amplification amounts for a second pair of polynucleotide expression
products of the CEACAM4 gene and LAMP] gene;
c) in a processing system:
i) retrieving the amplification amounts;
ii) determining an indicator by:
(1) determining a first derived biomarker value indicative of a ratio of
concentrations of the first pair of polynucleotide expression products by
determining a difference between the amplification amounts for the first
pair;
(2) determining a second derived biomarker value indicative of a ratio of
concentrations of the second pair of polynucleotide expression products by
determining a difference between the amplification amounts for the second
pair;
(3) determining the indicator by adding the first and second derived biomarker

values;
iii) retrieving previously determined first and second indicator references
from a
database, wherein the first and second indicator references are distributions
of
indicators determined for first and second groups of a reference population,

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 197 -
the first and second group consisting of individuals diagnosed with inSIRS
and ipSIRS respectively;
iv) comparing the indicator to the first and second indicator references;
v) using the results of the comparison to determine a probability of the
subject
being classified within the first or second group;
vi) generating a representation at least partially indicative of the indicator
and the
probability; and,
vii)providing the representation to a user to allow the user to assess the
likelihood
of a biological subject having at least one medical condition.
[0711] Additionally, a method can be provided for determining an indicator
used in assessing
a likelihood of a biological subject having a presence, absence, degree or
prognosis of at least
one medical condition, the method including:
a) determining a plurality of biomarker values, each biomarker value being
indicative of a value measured or derived for at least one corresponding
immune
system biomarker of the biological subject and being at least partially
indicative
of a concentration of the immune system biomarker in a sample taken from the
subject;
b) determining the indicator using a combination of the plurality of biomarker

values, wherein:
i) at least two biomarkers have a mutual correlation in respect of the at
least one
condition that lies within a mutual correlation range, the mutual correlation
range being between 0.9; and,
ii) the indicator has a performance value greater than or equal to a
performance
threshold representing the ability of the indicator to diagnose the presence,
absence, degree or prognosis of the at least one condition, the performance
threshold being indicative of an explained variance of at least 0.3.
[0712] Throughout this specification and claims which follow, unless the
context requires
otherwise, the word "comprise", and variations such as "comprises" or
"comprising", will be
understood to imply the inclusion of a stated integer or group of integers or
steps but not the
exclusion of any other integer or group of integers.

CA 02930925 2016-05-17
WO 2015/117204 PCT/AU2015/050043
- 198 -
[0713] Persons skilled in the art will appreciate that numerous variations and
modifications
will become apparent. All such variations and modifications which become
apparent to
persons skilled in the art, should be considered to fall within the spirit and
scope that the
invention broadly appearing before described.

TABLE 1
HGNC 5 Gene SEQ LRRC42 29 TIPRL 58
MRPS15 87
Name ID NO
C1orf175//TTC4 30 SFT2D2 59
RRAGC 88 o
PRKCZ 1 TM EM61 31 CACNA1 E 60
COL9A2 89


SKI 2 FPGT//TNNI3K 32 SMG7 61
TESK2 90 vi
1-


RER1 3 ACADM 33 OCLM 62
NRD1 91 --4
TAS1R1 4 SPATA1 34 RGS2 63
KTI12 92 .6.
VAM P3 5 EPHX4 35 ZC3H11A//RP11- 64
CC2D1B 93
74E24.2
AGTRAP 6 RPAP2 36
YIPF1 94
MFSD4 65
VPS13D 7 RPL5//SNORA66//S 37
JAK1 95
IL20 66
NORD21//FAM 69A
KLHDC7A 8
SLC35D1 96
RTCD1 38 RPS6KC1 67
NBL1//C1orf151 9
DIRAS3 97
SLC30A7 39 C1orf95 68
MDS2 10
ZZZ3 98
RN PC3//AMY2B 40 ARF1 69
P
RCAN 3 11
GNG5 99 o
r.,
CELSR2 41 GALNT2 70
.
LDLRAP1 12
ZN HIT6 100
AHCYL1 42 TN FRSF4 71
u,
MAN1C1 13
ODF2L 101
CEPT1//DRAM2 43 NADK 72
.
,
SH3BGRL3 14
SEP15 102 ,
,
CHIA 44 F1114100//C1orf86 73
7:; .
u,
,
DHDDS 15
BARH L2 103 z) ,
LIX1 L 45 GPR153 74
HCRTR1 16
GCLM 104
UPF0627 46 RERE 75
CCDC28B 17
CLCC1//GPSM2//C 105
MRPS21 47 SLC2A7 76
1orf62
LCK 18 TN FAI P8L2 48 SDHB 77
SORT1 106
ZN F362 19 SMCP 49 RN F186 78
SLC16A4 107
THRAP3 20 DCST1 50 DDOST 79
PHTF1 108
PPI E//CCDC25 21
1-d
RAG1AP1 51 GPN2 80
RSBN1 109 n
CAP1 22
1-3
C1orf182 52 RPA2 81
DEN N D2C//BCAS2 110 5;
CTPS 23
t.)
HAPLN 2 53 PEF1 82
CD58 111
C1orf84 24


NTRK1 54 PTP4A2 83
SPAG17//WDR3 112 vi
FAAH 25
-a-,
vi
CD1E 55 TRIM62 84
REG4//NBPF7 113
DM BX1 26
.6.
TOMM4OL//NR1I3 56 PHC2 85
RP11- 114 c,.)
CYP4B1 27
94I2.2//NBPF16//N
POU2F1 57 LSM10 86
BTF3L4 28
BPF11//NBPF15//N

BPF8//NBPF20//NB F5 140 TARBP1 168
GNLY 198
PF10//NBPF14//NB
PF1//L0C10028814
PIGC 141 CHM L 169
KCNIP3 199 0
n.)
2//NBPF12//KIAA1 KIAA0040 142 AKT3 170
CN N M4 200 o
1-,
245//L0C1002901 TOR1AIP2//TOR1AI 143 SMYD3 171
CN N M3 201 un
1-,
37 P1//IFRG15
AHCTF1 172
ZAP70 202 --.1
APH1A 115 STX6//KIAA1614 144
n.)
o
POGZ 116 EDEM3 145 OR1C1 173
LIPT1//MRPL30 203 .6.
TDRKH 117UCHL5 146 NCOA1 174
MAP4K4 204
THEM4 118 DEN N D1B 147 HADHB 175
IL1R2 205
S100A11 119 DDX59 148 ABHD1//PREB 176
IL1R1 206
CRNN 120 KIF21B 149 SPAST 177
IL18R1 207
SPRR2C 121ARL8A 150 SLC30A6//DDX50 178
POLR1B 208
S100Al2 122 CYB5R1 151 CRIPT 179
CHCHD5 209
P
S100A8 123 MYBPH 152 MSH2 180
IL1RN 210 2'
GATAD2B//PLI N 2 124 CHI3L1 153 FOXN2 181
PSD4 211 o
`,:,'
u,
DEN N D4B 125 PIK3C2B//LOC1001 154 CCDC104 182
DDX18 212
tv
"
.
1-
PBXIP1 126 30573 VRK2 183
INSIG2 213 c)
,
?
NUAK2 155 AHSA2//USP34 184
TM EM177//LOC100 214 ' ÷
'
PYGO2 127
1-
,
NUCKS1 156 OTX1 185
125918
SHC1 128
RALB 215
FAIM3 157 AFTPH 186
DCST2 129
PROC 216
PLXNA2 158 CEP68 187
GBA//G BAP 130
GPR17//L0C10029 217
SLC30A1 159 PLEK 188
ASH1L 131
1428//LIMS2
LPGAT1 160 ANXA4 189
IMP4 218
RIT1 132
ANGEL2 161
MEF2D 133 MXD1 190
FAM 123C 219
IV
n
RAB3GAP2//AURKA 162 NAGK 191
ACVR2A 220 1-3
AIM2 134
PS1//AURKA//SNO
5;
COPA 135 RA36B SMYD5//NOTO 192
MBD5 221
DEDD 136 TP53BP2 163 MTH FD2 193
LYPD6B 222
un
TADA1L 137 NVL 164 TTC31 194
SLC4A10 223
un
o
GPA33 138 TM EM63A 165 SEMA4F 195
UBR3 224 o
.6.
CD247 139 PARP1 166 TMSB10 196
HAT1 225
ITPKB 167 SH2D6 197
ITGA6 226

ZAK 227 PPM1G 257 MPP4 287
GYG1 317
OSBPL6 228 NLRC4 258 IN080D 288
SELT 318 0
PLEKHA3 229 CDC42EP3 259 KLF7 289
MED12L 319 w
o
1-,
ZC3H15 230 HNRPLL 260 FAM119A 290
RAP2B 320 vi
1-,
1-,
COL3A1 231 COX7A2L 261 NGEF 291
MYNN 321 --4
w
o
GLS 232 KCNG3 262 ARL4C 292
ABCF3 322 .6.
OBFC2A 233 CALM2//C2orf61 263 RAB17 293
VPS8 323
COQ10B 234 BCL11A 264 HDLBP 294
HRG 324
MARS2 235 XPO1 265 LRRN1 295
EIF4A2//SNORA4 325
CFLAR 236 NAT8B 266 SETD5 296
LPP 326
N0P58 237 DUSP11 267 IRAK2 297
CCDC50 327
FAM117B 238 MOGS 268 C3orf42 298
L0C152217 328
P
CYP20A1 239 SNRNP200 269 TSEN2 299
TADA3L 329 o
N,
u,
FASTKD2 240 SEMA4C 270 NR2C2//MRPS25
300 SEC13 330
u,
N,
PIKFYVE 241 MITD1 271 UBE2E1 301
TIMP4 331 u,
N,
tv
c,
,
C2orf62 242 IL1A 272 C3orf35 302
METTL6 332
,
u,
SLC11A1 243 SLC35F5 273 SNRK 303
DAZL//DAZ4//DAZ 333 ,
AGFG1 244 CCDC93 274 ZNF197 304

SATB1//TBC1D5
334
CHRNG 245 CLASP1 275 GNAI2 305
SCN10A
335
EIF4E2 246 SAP130 276 ALAS1 306
SEC22C
336
TRPM8 247 YSK4 277 PRKCD 307
ZDHHC3a
337
LRRFIP1 248 GTDC1 278 CACNA1D 308
ZDHHC3b
338
GAL3ST2 249 ORC4L 279 PXK 309
1-d
SLC6A20
339 n
TMEM18 250 NR4A2//F1146875 280 PTPRG 310
1-3
UQCRC1
340

LAPTM4A 251 DPP4 281 ATXN7 311
t.)
PRKAR2A
341
SF3B14 252 GALNT3 282 SLC35A5 312
1-,
IMPDH2
342 vi
TP53I3 253 SCN7A 283 SLC15A2 313
-a-,
CCDC71
343 vi
o
UNQ2999 254 FRZB 284 CCDC48 314
c,
UBA7
344 .6.
GPR113//SELI 255 STK17B 285 DNAJC13 315
CAMKV
345
MPV17 256 CLK1//PPIL3 286 CLDN18 316

WDR82 346 TMEM33//DCAF4L1 374 GPRIN3 403
GRPEL2 432
LMOD3 347 KIT 375 PPA2 404
MFAP3 433 0
FOXP1 348 ENAM 376 COL25A1 405
GABRA6 434 tµ.)
o


MORC1 349 FAM47E//STBD1 377 C4orf3 406
GABRA1 435 un
1-


ATG3 350 ENOPH1 378 QRFPR 407
DOCK2 436 --4
tµ.)
o
GSK3B//LOC10012 351 PDLIM5 379 MFSD8 408
RANBP17//USP12 437 .6.
9275 CCDC109B//HIGD1 380 MAP9 409
ERGIC1 438
HCLS1 352 A//CCDC13 PDGFC 410
ATP6V0E1//SNORA 439
KPNA1 353 EGF 381
74B
TKTL2 411
PTPLB 354 PCDH10 382
ZNF346 440
ACSL1 412
C3orf22 355 RAB33B 383
NSD1 441
SUB1//TMEM183A 413
RPN1 356 TMEM184C 384
CLPTM1L 442
CARD6 414
KIAA1257//ACAD9/ 357 RBM46 385
UGT3A1 443 P
/L0C100132731 MCCC2 415
o
N,
GRIA2 386
GDNF 444
FOXL2 358 TNP01 416
w
C4orf39 387
11C33 445
N,
MECOM 359 PDE8B 417
u,
KLHL2 388
hCG 2039148 446
PLD1 360 PAPD4 418
,
,
TLL1 389
MOCS2 447 tv .
,
GNB4 361 THBS4 419
u,
,
F11 390
SLC38A9 448 ,
,
MRPL47 362 FAM151B 420
SLBP 391
CCDC125 449
KLHL6 363 RASGRF2 421
HAUS3//POLN 392
ANKRA2 450
THPO 364 SNX2 422
PPARGC1A 393
HAPLN1 451
ETV5 365 LMNB1//PCIF1 423
TLR10 394
CCNH 452
BCL6//LOC100131 366 MEGF10 424
635 C4orf34 395
TMEM161B 453
LEAP2 425
1-d
ATP13A5 367 TXK 396
MBLAC2 454 n
TCF7 426
1-3
TMEM44 368 RPL21P44 397
MCTP1 455 5;
KDM3B 427
t.)
KIAA1530 369 KDR 398
TICAM2//TMED7//T 456
CXXC5 428
MED7-TICAM2 1¨

TACC3 370 RCHY1 399
un
SLC4A9 429
KIF3A 457 -,-:--,
CNO 371 CNOT6L 400
un
o
ANKHD1- 430
C5orf15 458 o
BST1 372 PLAC8 401
EIF4EBP3//ANKHD
SKP1 459
KLF3 373 HPSE 402 1//EIF4EBP3
KIAA0141 431
CXCL14 460

KLHL3 461 MAPK13 490 RXRB 520
C441208
CD14 462 PNPLA1 491 VPS52 521
GPR141 550 o
YIPF5 463 SFRS3 492 TCP11 522
STARD3NL 551 o

vi
LARS 464 CDKN1A 493 CLPS 523
POU6F2 552 1-


DCTN4 465 FOXP4 494 PGC 524
CDC2L5 553 --4
o
CCDC69 466 CUL9 495 ZNF318 525
ZMIZ2 554 .6.
ATOX1 467 RUNX2 496 YIPF3 526
UPP1 555
TIMD4 468 ZNF451 497 MRPL14 527
ZNF273 556
ADAM19 469 SOBP 498 PLA2G7 528
KCTD7//RABGEF1 557
SLIT3 470 C6orf182 499 PKHD1 529
RABGEF1//tcag7.9 558
RNF44 471 KIAA1919 500 IL17F 530
67//tcag7.951//KC
TD7//LOC1002933
DOK3 472 RWDD1 501 HTR1B 531
33 P
MGAT4B//SQSTM1 473 KPNA5 502 GABRR2 532
CCDC132 559 .
"
C5orf45//SQSTM1 474 TPD52L1 503 UBE231 533
PVRIG//PILRB//ST 560
.
AG3
"
RASGEF1C 475 ARG1 504 BACH2 534
PILRB//PVRIG//ST 561 "
t.)
,
MGAT1 476 RAB32 505 MCM9 535
AG3 .
,
(...)
.
IRF4 477 ARID1B 506 VNN1 536
C7orf51 562
,
,
HIVEP1 478 SLC22A3 507 IL2ORA 537
GNB2 563
E2F3 479 SERPINB1 508 F1127255 538
LRRC17 564
HIST1H4I 480 C6orf146 509 T 539
LRRN3 565
HIST1H2BM 481 GCM2 510 RPS6KA2 540
CFTR 566
MOG 482 ATXN1 511 HGC6.3 541
LSM8 567
LUC7L2
568
ZNRD1//NCRNA00 483 DCDC2//KAAG1 512 UNC84A//C7orf20 542
1-d
171
MGAM//LOC10012 569 n
HIST1H3I 513 SDK1 543
1-i
TRIM15 484
4692

HIST1H4L 514 ZDHHC4 544
GIMAP7 570 t.)
HCG27 485
GABBR1 515 C7orf26 545
INSIG1 571 1¨

BAT2//SNORA38 486
vi
RNA243 516 GLCCI1//tcag7.903
546 RBM33 572 -a-,
vi
CYP21A2 487
o
DDAH2 517 GPNMB 547
ICA1 573 =
.6.
ITPR3 488
c,.)
CLIC1 518 CCDC126 548
FAM126A 574
MAPK14 489
NEU1 519 WIPF3//ZNRF2//L0
549 HIBADH 575

TRIL 576 SGK196 604 RGP1//GBA2 633
FBXW5 663
SCRN1 577 UBE2V2 605 TGFBR1 634
C10orf18 664 o
ELMO1 578 F1146365 606 C9orf6//IKBKAP
635 FBX018 665 t.)
o


INHBA 579 SNTG1 607 IMAGE5303689 636
GATA3 666 vi
1-


CAMK2B 580 TRIM55 608 ATP6V1G1 637
CUGBP2 667 --4
t.)
o
NPC1L1 581 C8orf45 609 TLR4 638
VIM 668 .6.
DDC//L0C1001294 582 PREX2 610 SET 639
STAM 669
27 PLEKHF2 611 MRPL41 640
WAC 670
NSUN5//NSUN5B// 583
NSUN5C BAALCHF1110489 612 C9orf68 641
BAMBI 671
CLDN3 584 1TC35 613 HAUS6//SCARNA8 642
ZNF487//L0C4399 672
C7orf23//DMTF1 585 MTBP 614 KLHL9 643
11
ALOX5
673
SRI 586 ZHX2 615 C9orf82 644
WDFY4
674 P
BET1 587 RNF139 616 NDUFB6//DFFB 645
.
SRGN
675 N,
MCM7 588 TG 617 SIT1 646
w
CCDC109A
676 u,
N,
GATS 589 DENND3//C8orf60 618 FAM108B1 647
u,
FAM149B1//FAM14 677
ATXN7L1//RINT1// 590 TNFRSF1OD 619 TRPM6 648
9B2 c) ,
cn
,
EFCAB10
u,
TRIM35 620 FRMD3 649
MINPP1 678 1 ,
KIAA1549 591
,
,
GSR 621 5LC28A3 650
PTEN//PTENP1 679
5LC37A3 592
WHSC1L1 622 BICD2 651
ENTPD1//C10orf13 680
SMARCD3 593
1
PCMTD1//PXDNL 623 C9orf84 652
MLL3//BAGE2 594
ABCC2 681
NCOA2 624 AKNA 653
CLN8 595
SFXN2 682
TRAM1//L0C28619 625 MEGF9 654
MSRA 596
SHOC2 683
0 C5 655
1-d
PIWIL2 597 RUNX1T1 626
ACSL5 684 n
GOLGA1//SCAI 656
1-3
NEFM//L0C100129 598 EXT1 627
BCCIP//DHX32 685 5;
717 SH2D3C 657
DDEF1IT1 628
FAM188A 686 t.)
EPHX2 599 FAM102A 658


CDC37L1 629
CUBN 687 vi
LEPROTL1 600 F1110232 659
-,-:--,
UBE2R2 630
SVIL//hCG 178349 688 vi
MAK16//C8orf41 601 ASB6 660
4 o
c'
UBAP1//KIF24 631
.6.
AP3M2 602 BAT2L 661
FAM13C//PHYHIPL 689 c,.)
GALT 632
FNTA 603 EDF1 662
ATAD1 690

ANKRD22 691 C11orf30 721 CCDC82 751
PXMP2//PGAM5 780
F1134077 692 C11orf82 722 K1AA1826 752
DCP1B 781 o
tµ.)
COX15 693 TMEM126B 723 MPZL3 753
SLC2A3//SLC2A14 782 o


un
ERLIN1 694 C11orf73 724 MPZL2 754
C3AR1 783 1-


ACTR1A 695 PIWIL4 725 H2AFX 755
PLBD1 784 --4
tµ.)
o
ABLIM1 696 L0C100132686 726 SIAE 756
TM7SF3 785 .6.
RAB11FIP2 697 PAFAH1B2 727 ZBTB44 757
ASB8//PHB 786
C10orf84 698 UBE4A 728 HSN2 758
LMBR1L 787
PRDX3 699 TRAPPC4 729 ADIPOR2 759
FMNL3//PRPF4OB 788
C10orf119 700 SC5DL 730 NCAPD2//SCARNA1 760
AAAS 789
NSMCE4A 701 VWA5A//0R10D1P 731 0//FADS1
NFE2 790
PTPN6 761
TALD01//INTS8 702 STT3A 732
GPR84 791
CLEC4D 762
P
TNNT3 703 VPS26B 733
CD63 792 o
CDKN1B 763
N,
w
w
FXC1 704 TRIM21 734
SARNP//DNAJC14 793 .
GOLT1B 764
u,
PDE3B 705 ZBED5 735
NACA 794 N,
FAR2 765
tv .
,
DNAJC24 706 SAAL1 736
CDK4//TSPAN31 795 .
,
FGD4 766
v, .
u,
PTPRJHOR4B1 707 FANCF 737
TMBIM4//LOC1001 796
,
TMEM106C 767
33322 ,
C11orf31 708 LIN7C 738
TMBIM6 768
IL22 797
TMEM109 709 PHF21A 739
C12orf62 769
LIN7A 798
CD6 710 CUGBP1 740
PRR13//PCBP2 770
HAL 799
CD5 711 OSBP 741
DGKA 771
APPL2 800
TMEM138 712 CYBASC3 742
COQ10A 772
GLTP 801
POLR2G 713 TUT1 743
1-d
TSPAN31 773
GIT2 802 n
TMEM179B 714 SLC25A45 744
1-3
CDK4/MARCH9/C3 774
VPS29 803 5;
NAT11 715 LTBP3 745 HC4
t.)
PPTC7
804
OTUB1 716 EIF1AD 746 LEMD3 775


DDX54//CCDC42B 805
un
RBM14//RBM4 717 GAB2 747 IRAK3 776
-,-:--,
SLC24A6
806 un
o
AIP 718 CREBZF 748 TMTC3 777
o
SDS
807 .6.
PPFIA1 719 PICALM 749 ACTR6 778
RBM19
808
IL18BP//NUMA1 720 SLC36A4 750 TCTN1 779

MED13L 809 ZMYM2 839 ELF1 867
HIF1A 896
C12orf49 810 SPATA13//C1QTNF 840 LCP1 868
SYNE2 897 o
9
tµ.)
FBX021 811 KPNA3 869
EXD2 898 o
NUPL1 841


un
WSB2 812 C13orf1 870
SLC39A9 899 1¨

PAN3//EEF1A1//CH 842


TAOK3 813 CHD2 DLEU2//DLEU2L
871 SFRS5 900 --4
t.)
o
CIT 814 ALOX5AP 843 GUCY1B2 872
PCNX 901 .6.
RAB35 815 EEF1DP3 844 INTS6 873
SIPA1L1//SNORD5 902
RPLPO 816 KL 845 DACH1 874
6B//L0C145474//L
0C283567
PXN 817 UFM1 846 TBC1D4 875
YLPM1 903
TRIAP1 818 NARG1L 847 EDNRB 876
BATF 904
SFRS9 819 ITM2B 848 UGGT2 877
FLVCR2//RPS24 905
POP5 820 FNDC3A 849 GPR183 878
GPR65 906 P
UNQ1887 821 CDADC1 850 LIG4 879
TDP1 907 .
N,
.
C12orf43 822 ARL11 851 ANKRD10 880
EVL 908 w
ND
ANAPC5 823 LMO7 852 RASA3 881
ZNF839 909 u,
''
tv
,
KDM2B 824 DNAJC3 853 RNASE2//L0C6433 882
TDRD9 910 .
,
MORN3 825 TM9SF2 854 32
INF2
911
,
RPGRIP1 883
,
TMEM120B//RHOF 826 CLYBL 855
PLD4 912
IRF9 884
L0C338799 827 PCCA 856
MTA1//L0C647310 913
TSSK4 885
DIABLO//B3GNT4 828 ABHD13 857
//L0C100128343
C14orf21 886
NDRG2 914
VPS33A 829 LAMP1 858
SCFD1 887
DAD1//OR631 915
CLIP1 830 TMC03 859
FANCM 888
SLC7A8 916
PITPNM2 831 UPF3A 860
Iv
ABHD12B 889
IP04 917 n
EIF2B1 832 ZMYM5//ZMYM2 861
1-3
PTGDR 890
TM9SF1 918 5;
CCDC92 833 ZDHHC20//L0C728 862
t.)
099 FBX034//KIAA083 891
ADCY4 919
NCOR2 834 1

PARP4 863
RIPK3 920 un
DHX37 835 C14orf101 892
-,-:--,
MTMR6//L0C64648 864
EAPP 921 un
o,
DDX51 836 2 ACTR10 893
o
.6.
POLE 837 HSPH1 865 ARID4A 894
BAZ1A 922 c,.)
GOLGA3 838 N4BP2L2//CG030 866 JKAMP 895
NFKBIA 923

SEC23A 924 CHP 954 MEF2A//LYSMD4
981 MTHFS 1008
C14orf104 925 3M3D7- 955 NIPA2//CYFIP1
982 ST20//C15orf37 1009 o
PLA2G4B//3M3D7//
tµ.)
C14orf138 926 HERC2//HERC2P2//
983 TMC3 1010 o
PLA2G4B

HERC2P3//L0C440
un
AP3B2
1011
SOS2 927 HAUS2 956 248
1-


NIN 928 C15orf63//SERF2 957 MTMR10//MTMR15 984
C15orf40 1012 --4
tµ.)
o
PYGL 929 B2M 958 C15orf24 985
WDR73 1013 .6.
CNIH 930 TRIM69 959 SLC12A6 986
NTRK3 1014
DHRS7 931 PLDN 960 LPCAT4 987
DET1 1015
WDR89 932 SQRDL 961 1N080 988
TM2D3 1016
ACTN1 933 GALK2 962 01P5 989
WDR90 1017
NUMB 934 USP8 963 ZFP106 990
RHOT2//FBXL16 1018
C14orf43 935 GLDN 964 CDAN1 991
TMEM204 1019
P
ABCD4 936 MAPK6 965 SPG11//ISLR 992
CRAMP1L//HN1L 1020 o
N)
KIAA0317 937 LACTB 966 SPPL2A 993
MAPK8IP3 1021
o
r.,
NEK9 938 RAB8B 967 GNB5//L0C100129 994
TBL3 1022 u,
''
ANGELI. 939 APH1B 968 973
TSC2tv
1023
,
,
MY05A 995
---.1 0
u,
SPTLC2 940 USP3//LOC100130 969
KCTD5//PR00461// 1024 ,
SERPINA6 941 855 ARPP19 996
PDPK1 ,
SNX1 970 RAB27A 997
CLUAP1 1025
DICER1 942
LBXCOR1//PIAS1// 971 CCPG1//PIGB//DYX
998 DNASE1 1026
BCL11B 943 CALML4 1C1
DNAJA3 1027
ANKRD9 944 NE01 972 BNIP2 999
CP110
1028
PPP1R13B 945 MPI 973 CA12
1000
C16orf62
1029
AKT1 946 FBX022//FBX0220 974 FAM96A
1001 1-d
LYRM1
1030 n
BRF1 947 S KIAA0101//CSNK1
1002 1-3
RCN2 975
METTL9 1031
TUBGCP5 948 G1
5;
t.)
FAH 976 TLE3
1003 EEF2K 1032
SNRPN * 949
1L16 977 977 PARP6
1004 POLR3E 1033 un
APBA2 950
-,-:--,
ABHD2 978 NPTN
1005 PLK1 1034 un
o
MTMR15//MTMR10 951
c'
SLCO3A1 979 MAN2C1
1006 PRKCB 1035 .6.
RYR3 952
MCTP2 980 IMP3
1007 IL21R//L0C283888 1036
BAHD1 953

SULT1A2//SULT1A 1037 KLHL36 1065 COG7
1092 VAC14//L0C10013 1120
1
0894
K1AA0182 1066 GGA2
1093 0
ATXN2L 1038
AP1G1 1121 r..)
BAN P//RUN DC2C 1067 NSMCE1
1094 o
LAT II 1039
WDR59 1122 1¨

un
TRAPPC2L 1068 GTF3C1
1095
KIF22 1040
CTRB2//CTRB1 1123 1-


SPG7 1069 CCDC101//L0C388
1096
MAZ 1041 242
TAF1C//ADAD2 1124 o
.6.
CDK10 1070
CORO1A//L0C6067 1042 C16orf54
1097 FBX031 1125
24 TCF25 1071
KCTD13
1098 ZCCHC14 1126
ITGAL 1043 AFG3L1 1072
SEPT1
1099 FAM 38A 1127
SRCAP//SNORA30 1044 LUC7L 1073
ZN F764//ZN F747
1100 CEN PBD1 1128
ZN F646//ZN F668 1045 AXIN1 1074
C16orf58//LOC100 1101
TIMM22 1129
C16orf67 1046 3M3D8 1075 128371
RPA1
1130
TM EM188 1047 LMF1 1076 ITFG1
1102
DPH1//0VCA2
1131 P
LPCAT2 1048 UN KL 1077 ABCC11//LONP2
1103 .
SGSM2
1132 N,
.
CETP 1049 UN KL 1078 N U DT21
1104 L.
ARRB2
1133 `,:,'
CKLF 1050 CLCN7 1079 BBS2//0GFOD1
1105 u,
ND
L0C100130950
1134 t,) .
CMTM1//CKLF 1051 MRPS34 1080 CSNK2A2
1106 ,
0.,
DNAH2
1135 00 ,
.
TM EM208 1052 RN PS1 1081 GOT2
1107 1 u,
,
PIGL
1136 ,
,
CTCF 1053 NLRC3 1082 FAM96B
1108
TRPV2
1137
THAP11 1054 TRAP1//DNASE1 1083 FHOD1//SLC9A5
1109
MPRIP
1138
NUTF2 1055 ADCY9 1084 ATP6V0D1//LOC10
1110
0132855
DRG2 1139
EDC4 1056 COR07 1085
GFOD2
1111 ALKBH5//F1113773 1140
SLC7A6//SLC7A60 1057 C16orf72 1086
S SLC12A4 1112 SMCR7 1141
RRN3//L0C653390 1087
PRMT7 1058 DPEP3
1113 WSB1 1142 n
//L0C730092//LOC
1-3
SNTB2 1059 100131998 DPEP2
1114 TAOK1 1143 5;
VPS4A 1060 XYLT1//LYRM2//ZC 1088 CHTF8//HAS3
1115 CPD 1144 t,.)
3H11A
DDX19B//DDX19A 1061 DCUN1D3//LYRM 1 1089 COG8//PDF
1116 SUZ12P 1145 1¨

un
'a
CHST4 1062 IGSF6//METTL9 1090 TERF2
1117 RN F135 1146 un
=
o
HP//HPR 1063 CDR2//RRN3//LOC 1091 AARS
1118 ZN F830 1147 .6.
PLCG2 1064 100131998//LOC6 ST3GAL2
1119 TAF15 1148
53390

GGNBP2 1149 TM EM104 1177 DERL2
1206 NR1D1//THRA 1232
LASP1 1150 MRPS7 1178 NLRP1//L0C72839
1207 CCR7 1233 0
2
tµ.)
PSM D3 1151 KIAA0195 1179
STAT5B//STAT5A 1234 o
ASGR2
1208 1¨

un
CDC6 1152 TSEN54 1180
FAM 134C 1235
NEURL4//GPS2//D 1209
1-


NBR2 1153 LLGL2 1181 4S234E
VAT1 1236 --4
t.)
o
TM UB2 1154 L0C100134934//C 1182 ZBTB4
1210 DUSP3 1237 .6.
DK3
MGC57346//C17orf 1155 TP53
1211 C17orf65//ASB16 1238
69 MFSD11 1183
VAM P2
1212 UBTF 1239
NSF//L0C728806 1156 SEPT9 1184
PIK3R5
1213 GPATC H8 1240
GOSR2 1157 TN RC6C 1185
ELAC2
1214 MAP3K14//LOC100 1241
NPEPPS//TBC1D3F/ 1158 TMC8 1186
/LOC440434 NCOR1//C200rf191
1215 133991
ENGASE 1187 //L0C100131704
OSBPL7 1242
KPN B1 1159
RPTOR 1188 ZN F287
1216 SLC35B1 1243 P
CDK5RAP3 1160
GPS1 1189 TOM1L2//L0C2463
1217 TOB1 1244 2
ATP5G1 1161 15
FN3KRP 1190
COX11//TOM1L1 1245 2
UBE2Z 1162 GRAP//SNORD3B- 1218
u,
TBCD 1191 1//SNORD3B-
VEZF1 1246 (,) r.,
o
XYLT2//LOC100130 1163
,
580 GEMIN4 1192 2//L0C400581
SFRS1//F1144342 1247 s:) ,
u,
ALDOC
1219 1 ,
NOG 1164 GLOD4 1193
SEPT4 1248 ,
,
SDF2
1220
DGKE 1165 SLC43A2 1194
MED13//LOC10012 1249
RAB34
1221 9112
AKAP1 1166 PRPF8 1195
PHF12
1222 LIMD2//MAP3K3 1250
TM EM49//CLTC//MI 1167 SMG6//C17orf6 1196
R21 N U FIP2 1223 STRADA 1251
METT10D//L0C284 1197
CLTC 1168
009 OMG
1224 FTS33 1252
CA4 1169 SHPK 1198 EVI2B
1225 CD79B 1253 1-d
C17orf64 1170 TAXI BP3 1199 C17orf66//RSL24D
1226 ICAM 2 1254 n
DCAF7 1171 P2RX5 1200 1
ERN 1 1255 5;
SYNRG//LOC10013 1227
t.)
PITPNC1 1172 MYBBP1A//SPNS2 1201 1822
TEX2 1256 1¨

un
NOL11//SNORA38B 1173 PELP1 1202 PLXDC1
1228 LRRC37A3//LRRC3 1257 -,-:--,
un
MAP2K6 1174 PFN1 1203 CACN B1
1229 7A2//LRRC37A//AR o
o
L17P1//LRRC37A4/
.6.
COG1 1175 ZN F232 1204 PGAP3
1230 /L0C100294335//L c,.)
CD300A 1176 DHX33 1205 MED24
1231 00644397

GNA13 1258 AFG3L2 1286 IL27RA
1314 CD177 1342
WIPI1//ARSG 1259 ABHD3 1287 KLF2
1315 ZNF230//ZNF222 1343 0
FAM20A 1260 OSBPL1A 1288 SIN3B
1316 VASP 1344 tµ.)
o


NAT9 1261 CDH2 1289 DDA1
1317 GRWD1 1345 un
1-


GGA3 1262 DSC1 1290 GTPBP3
1318 FLT3LG 1346 --4
tµ.)
o
H3F3B//H3F3C 1263 PSTPIP2 1291 FAM129C
1319 ZNF175 1347 .6.
EXOC7 1264 C18orf32 1292 FCH01
1320 NCRNA00085 1348
SFRS2 1265 MBD2//SNORA37 1293 ARRDC2
1321 PPP2R1A 1349
TMC6//LOC100131 1266 PIGN 1294 IFI30
1322 ZNF808//ZNF578// 1350
096
ZNF611
TMX3 1295 C19orf60
1323
USP36 1267
LENG8 1351
PQLC1 1296 CRTC1//MAML2
1324
CD7 1268
FCAR 1352
GZMM 1297 RFXANK//MEF2B//L
1325
RAB31 1269 00729991
RPL28 1353 P
ARID3A 1298
0
VAPA 1270 ZNF101
1326 U2AF2 1354 N,
CIRBP 1299
w
0
SEH1L 1271 ZNF738
1327 L0C100288114//M 1355 .
N,
DAZAP1 1300
GC9913 u,
N,
HQ0644/PR00644 1272 ZNF257//ZNF492//
1328 tv .
SPPL2B 1301 ZNF99//ZNF98//L0
ZFP28 1356 ,
,
RNMT 1273
8 .
NFIC 1302 C646864
ZNF460 1357 u,
1
,
-JRNF138 1274
C19orf2 1329 ,
VAV1 1303
ZNF549 1358
GALNT1 1275 K1AA0355//F11213
1330
ARHGEF18//LOC10 1304 ZNF211 1359
69
ELP2 1276 0128573
USF2
1331 ZNF587//ZNF417 1360
PIK3C3 1277 STXBP2//L0C5543 1305
SLC14A2 1278 63//L0C10013180 TMEM147
1332 ZNF274 1361
1 LIN37//PSENEN
1333 ZNF544 1362
ME2 1279 C19orf59 1306
C19orf55
1334 ZNF8 1363 1-d
SERPINB2//SERPIN 1280 ZNF317 1307
n
B10 TBCB//POLR2I
1335 TRIM28 1364 1-3
ILF3 1308
5;
ZNF407 1281 ZNF382
1336 C19orf6 1365
t.)
SMARCA4 1309
ZNF236 1282 ZNF568
1337 C19orf34 1366 1¨

PRKCSH 1310
un
NFATC1//LOC1001 1283 ZNF420
1338 GNG7 1367 -,-:--,
un
27994 IER2 1311
=
ZNF383
1339 AES 1368 o
ENOSF1//TYMS 1284 CCDC130 1312
.6.
CCDC97
1340 EEF2//SNORD37 1369
MYOM1 1285 DCAF15 1313
ZNF574
1341 PLIN5//LRG1 1370

PLIN3 1371 ZNF529 1399 CSRP2BP
1429 C200rf177 1458
PTPRS 1372 ZNF461 1400 SEC23B
1430 CDH26 1459 0
SAFB2//SAFB 1373 ZNF607 1401 SLC24A3
1431 C200rf197 1460 tµ.)
o


RANBP3 1374 YIF1B 1402 HCK
1432 L0C284757 1461 un
1-


GTF2F1//LOC1001 1375 PRR13 1403 ASXL1
1433 ARFGAP1 1462 --4
tµ.)
30856
o
CEACAM4 1404 ACSS2
1434 PRPF6 1463 .6.
XAB2 1376
PLAUR 1405 C200rf4
1435 NSFL1C 1464
ELAVL1 1377
TRAPPC6A 1406 TGIF2
1436 SIRPD 1465
ADAMTS10 1378
ERCC1//CD3EAP 1407 C200rf24//SLA2
1437 SIRPG//SIRPA 1466
FBXL12 1379
RTN2 1408 RPN2//EEF1A2
1438 RNF24 1467
DNMT1 1380
SYMPK 1409 CTNNBL1
1439 RASSF2 1468
TYK2 1381
PGLYRP1 1410 ACTR5
1440 TMX4 1469
KEAP1 1382
P
NOSIP 1411 PPP1R16B
1441 JAG1 1470 o
KRI1 1383
N,
PNKP 1412 DHX35
1442 C200rf74 1471 w
o
TMEM205//hCG 29 1384
.
N,
977 NKG7 1413 PLCG1
1443 C200rf3 1472 u,
N,
.
ZNF563 1385 FPR1 1414 MYBL2
1444 C200rf112 tv
1473

,
,
,
.
MAN2B1//MORG1 1386 ZNF28 1415 SYS1//SYS1-
1445 CDK5RAP1 1474 , u,
,
,
C19orf56 1387 OSCAR 1416 DBNDD2//DBNDD2
AHCY 1475 ,
DNTTIP1
1446
DHPS 1388 MBOAT7 1417
GGT7 1476
CTSA
1447
TNP02//SNORD41 1389 LILRA5 1418
EDEM2 1477
MMP9//L0C100128 1448
LPHN1 1390 LILRA4 1419 028
RBM39//L0C64316 1478
NDUFB7 1391 ZNF550//ZNF549 1420 DDX27
1449 7
BLCAP
1479
AKAP8 1392 ZNF416 1421 SLC9A8
1450 1-d
SERINC3//TTPAL
1480 n
AKAP8L 1393 ZNF256 1422 RNF114
1451 1-3
ZNF335
1481 5;
CHERP//C19orf44// 1394 ZNF329 1423 PTPN1
1452
t.)
CALR3
ELMO2 1482
FAM110A 1424 TSHZ2
1453
INSL3//JAK3 1395
B4GALT5 1483 1¨

un
ITPA 1425 PFDN4
1454 -c-:--,
IL12RB1 1396
DPM1 1484 un
o
CDC25B 1426 CSTF1
1455 o
UPK1A 1397
ZFP64 1485 .6.
CDS2 1427 CASS4
1456
TYROBP 1398
ZNF217 1486
CRLS1 1428 GNAS
1457

CTSZ 1487 CSTB 1516 RP3-402G11.5
1544 MOSPD2 1572
SYCP2 1488 UBE2G2//SUM03 1517 SAPS2
1545 BMX//HNRPDL 1573 o
tµ.)
PSMA7 1489 PTTG1IP 1518 NCAPH2
1546 PDHA1 1574 o


un
DIDO1 1490 POFUT2 1519 BID
1547 YY2 1575 1-


YTHDF1 1491 MCM3AP 1520 SLC25A1
1548 PDK3 1576 --4
tµ.)
o
CHODL 1492 IL17RA//CECR7 1521 KLHL22//KRT18 1549
GKHGK3P//FTL//L 1577 .6.
00652904
BACH1 1493 C22orf37 1522 PI4KA//PI4KAP1//P
1550
I4KAP2//L0C10029
CXorf59 1578
C21orf41//BACH1 1494 LZTR1 1523
3141
ATP6AP2 1579
IL1ORB 1495 PPIL2//YPEL1 1524 MAPK1
1551
USP9X//USP9Y
1580
IFNAR1 1496 CYTSA 1525 ZNF70
1552 RP2 1581
IFNGR2 1497 SNRPD3//C22orf13 1526 TPST2
1553 USP11 1582
SON 1498 NF2 1527 SF3A1//CCDC157 1554
RBM3 1583 P
MORC3//DOPEY2 1499 LIMK2 1528 PES1
1555 0
r.,
FTS31
1584 .
DYRK1A 1500 SLC5A1 1529 PIK3IP1
1556 0
WAS
1585
KCN315 1501 MCM5 1530 PATZ1
1557 r.,
PLP2
1586 o
,
ETS2 1502 NCF4 1531 C22orf30
1558 .
,
TSPYL2//GPR173
1587
RRP1B 1503 GGA1 1532 IL2RB
1559 ,
,
MAGED2
1588 ..,
PFKL 1504 SH3BP1//PDXP 1533 CSNK1E//L0C4009
1560 UBQLN2 1589
TRPM2 1505 POLR2F//LOC1001 1534 27
31530 UNC84B
1561 NLGN3 1590
ADARB1 1506
APOBEC3A//APOBE 1535 CBX7//L0C100128
1562 ACRC 1591
SAMSN1//L0C3888 1507 C3B 400
UPRT 1592
13 APOBEC3D 1536 RPS19BP1
1563
N6AMT1 1508
CXorf26 1593
ATF4 1537 MKL1//KIAA1659
1564 1-d
SYN31 1509
ATP7A 1594 n
CACNA1I 1538 RANGAP1
1565 1-3
TMEM5OB 1510
DIAPH2 1595 5;
ZC3H7B 1539 TCF20
1566
KCNE1 1511
CSTF2//RAD21 1596 t.)
CCDC134 1540 LDOC1L
1567 1¨

PRDM15 1512
ARMCX3 1597 un
TSPO 1541 UNQ6126
1568
C2CD2 1513
ARMCX5 1598 un
o
NUP50 1542 TUBGCP6
1569
WDR4 1514
GPRASP1 1599 .6.
TBC1D22A//LOC10 1543 SBF1//SBF1P1
1570
U2AF1 1515 0289878
TMEM31 1600
MSL3
1571

TBC1D8B 1601 /SNORA56 GLA
1628 DNASE1L1//RPL10 1641
MID2 1602 ARSD 1615 MORF4L2
1629 SH3KBP1 1642
0
DOCK11 1603 KALI. 1616 PSM D10
1630 Mitochondrial1 1643 n.)
o


LON RF3 1604 CTPS2 1617 ACSL4
1631 Mitochondrial2 1644 un
1-


UBE2A 1605 RPS6KA3 1618 LAMP2
1632 CCN L2 1645 -4
n.)
o
SH2D1A 1606 BCOR 1619 CU L4B
1633 IN PP5B 1646 .6.
OCRL 1607 MA0B//NAT13 1620 ODZ1
1634 TLR5 1647
SLC25A14 1608 ZN F41 1621 ELF4
1635 ADRB3//GOT1 L1 1648
HPRT1 1609 OTUD5 1622 RAP2C
1636 NOC2L//SAMD11// 1649
CD4OLG 1610 KCN D1 1623 FAM127B//FAM127
1637 LOC401010
AFF2 1611 ZMYM3 1624 C//FAM 127A
SHFM1 1650
TM EM185A
1638
SSR4//IDH3G 1612 MAGT1 1625
ARD1A
1639 P
FAM 50A 1613 BRWD3 1626
o
IRAK1
1640 "
DKC1//SNORA36A/ 1614 TRMT2B 1627
o
r.,
6//SNORD115-9//SNORD116- 8//SNORD115-
3//SNORD115- u,
r.,
t.) .
HUGO Gene Nomenclature 11//SNORD116-12//SNORD116- 38//SNORD115-
41//SNORD115-
,
Committee 13//SNORD116-28//SNORD116- 22//SNORD115-
44//SNORD116- La
u,
.
,
4//SN OR D64//PARV/SN OR D109A// 1//SNORD115-
17//SNORD115- ,
,
T1 Synonymous with SNORD109B//SNORD116- 18//SNORD115-
19//SNORD115-
SPNS1//N PI PL2//L0C728741//L0C73 6//SNORD116-
3//SNORD116- 20//SNORD116@
0153//N PI PL3//SPI N1//LOC728888// 9//SNORD115-13//SNORD115-
LOC100289169//LOC728734//LOC72 1//SNORD115-14//SNORD115-
9602//LOC100288442//LOC1002883 15//SNORD115-21//SNORD115-
32 10//SNORD115-7//SNORD115-
16//SNORD115-40//SNORD115-
1-d
* Synonymous with 42//SNORD115-11//SNORD115-
n
SNURFHIPW//SNORD116- 29//SNORD115-34//SNORD115-
5;
16//SNORD116-18//SNORD116- 36//SNORD115-4//SNORD115-
t.)
21//SNORD116-22//SNORD116- 43//HBII-52-24//SNORD116-


un
17//SNORD116-19//PAR5//PAR- 5//SNORD116-7//SNORD115-
-a 5
un
SN//SNORD116-2//SNORD116- 26//SNORD115-30//SNORD116-
=
o
25//SNORD116- 15//SNORD116-8//SNORD115-
.6.
26//SNORD107//SNORD115- 2//SNORD115-39//SNORD116-
12//SNORD115-5//SNORD115- 14//SNORD116-20//SNORD115-

TABLE 2
HGNC 5 Gene SEQ CYP4B1 1677 NTRK1
1704 PEF1 1732
Name ID NO BTF3L4 1678 CD1E
1705 PTP4A2 1733
PRKCZ 1651 LRRC42 1679 TOMM40L//NR1I3 1706
TRIM62 1734 0
SKI 1652 C1orf175//TTC4 1680 POU2F1
1707 PHC2 1735 o


vi
RER1 1653 TMEM61 1681 TIPRL
1708 LSM10 1736 1-
1-
--4
TAS1R1 1654 FPGT//TNNI3K 1682 SFT2D2
1709 MRPS15 1737 t,.)
o
.6.
VAMP3 1655 ACADM 1683 CACNA1E
1710 RRAGC 1738
AGTRAP 1656 SPATA1 1684 SMG7
1711 COL9A2 1739
VPS13D 1657 EPHX4 1685 OCLM
1712 TESK2 1740
KLHDC7A 1658 RPAP2 1686 RGS2
1713 NRD1 1741
NBL1//C1orf151 1659 RPL5//SNORA66// 1687 ZC3H11A//RP11- 1714
KTI12 1742
SNORD21//FAM69 74E24.2
MDS2 1660 A MFSD4
1715 CC2D1B 1743
RCAN3 1661 RTCD1 1688
YIPF1 1744 P
IL20
1716 .
LDLRAP1 1662 SLC30A7 1689
JAK1 1745 "
RPS6KC1
1717
MAN1C1 1663 RNPC3//AMY2B 1690
SLC35D1 1746 .
r.,
C1orf95
1718 u,
SH3BGRL3 1664 CELSR2 1691
DIRAS3 1747
ARF1
1719 tv ,
,
DHDDS 1665 AHCYL1 1692
ZZZ3 1748
GALNT2
1720 -Z: .
u,
,
HCRTR1 1666 CEPT1//DRAM2 1693
GNG5 1749 ' ,
_.]
TNFRSF4
1721
CCDC28B 1667 CHIA 1694
ZNHIT6 1750
NADK
1722
LCK 1668 LIX1L 1695
ODF2L 1751
F1114100//C1orf8 1723
ZNF362 1669 UPF0627 1696 6
SEP15 1752
THRAP3 1670 MRPS21 1697 GPR153
1724 BARHL2 1753
PPIE//CCDC25 1671 TNFAIP8L2 1698 RERE
1725 GCLM 1754
CAP1 1672 SMCP 1699 SLC2A7
1726 CLCC1//GPSM2//C 1755 1-d
n
CTPS 1673 DCST1 1700 SDHB
1727 1orf62 1-3
SORT1
1756 5;
C1orf84 1674 RAG1AP1 1701 RNF186
1728 t.)
SLC16A4
1757
FAAH 1675 C1orf182 1702 DDOST
1729 1¨

vi
PHTF1
1758
DMBX1 1676 HAPLN2 1703 GPN2
1730 -a-,
vi
RSBN1
1759
RPA2
1731 o
4=,
W

m
7r 6081
11VDd1
o
o 17981
11V?:1 18LT Illl:1
in 9E81 89d9D
= 816SZ TO 8081
TVOED-IS
in SE8T HdldV
08L1 -I T HSV
L081
ZVNX
,-i 981 OTDOV/LL T 149141
-Id
o
el Z981 ZDISNI 1 6LLT d V9
DHVEID781 TX10 9081 14IVd
-e
8LLT Z1SDa
T98T 8 TXCla E8T VEdS1-1//ZVSHV
E-1 S081
TS>01-1N
c.) ZE8T ZMA
LLLT TDHIS
Po 0981 KISd
17081 z>ivnN
6581 N?:IT-II 181 VOIDIODD
ELSOET 9LLT ZODAd
SCHDHID 0E81 ZNX0d 081 00TDO-
IHEIZDENId SLLT TdIX9d
8581
6Z81 ZHS14 Z081
TlEIHD 17LL1
EIKINN9C1
LS8T 9 TWIOd
1081
Hd9A14 ELL T ZNI-IdHEIZCIV1VD
9581 1811I 8Z81 ldnID
LZ81 OSX1010//9VOED-IS 0081 T?:ISEIAD ZLLT 8VOOTS
SS8T T?:I T-II
9Z81 1SVdS 66L1
V8-1?:IV ILL T ZTVOOTS
`,--1 17S81 Z?:I T-II
86L1
9 TZdI>1 DDP:IdS
' ES81 17>117dV14 SZ8T 19?:IdHIGH9V
L6L1
6SXCla OLLT NN?:ID
,cs'i C7,1 ZS8T 0E-1&:114//T1dI-1 17Z81
9HICIVH
,0
TS8T OLdVZ EZ8T TVODN 96L1
9 TaNN9C1 69L1 T TVOOTS
.
S6LT
S-11-1DC1 89L1 171491-11
OS81 14NND ZZ8T TDT?:10
'8'
176L 1
149a9 L9L1 H>P:11:11
0 61781 1714NND 1Z81 1d1DHV
EdIND>1 OZ81 EGANS E6L T VT 9 TVVI>1//9X1S
99L1 ZDOd
81781
STD?:IdIi/TdI
L1781 A-IND 6181 El>1V
Z6LT V1?:101//ZdIV1?:101 S9LT VT HdV
LE106
91781 910ZHIS 8181 114HD 16L1
01700VVI>1 ZOOTDOV/SVZ TVV
S1781 OT9S141 181 1d9lV1 06L1
DDId INHZ1dd9NHZ1718
1717 8 1 di7V149S 9181 9>idlI 68L1
Sd 8Z001DO1//1dd9N/it 1dd9 NHOTdd9N
1781 T E D11 S181 Td?:IVd
88L1 L17= H0Zdd9 NH8dd9 N
7r
1
HS Tdd9NHT Tdd9N
el= Z1781 Zad H114 7181 VE9149141
L8L1 EVdD
N
//91dd9NHZ.ZI176
,-i
,-i 11781 010 N HSCIA14 S 181 lAN
98L1 TIVCIV1 179L 1 -11d
,-i 01781 >19VN Z181 ZdEIESd1
S8L1 la:19a E9LT Ldd9N/i1799?:1
o
o 6E81 TaXl4
99EV?10
NS/NM nwir SdV 178L 1 Vd0D Z9LT
alaMHLTDVdS
8E81 17VXNV 1181
>1?:Inviizdvoz9n E8LT ZNIV 19L1 890D
LE8T >19-Id 0181 Z199NV Z8LT
aZd914 09L1 ZSVD9HDZCINN9C1

PROC 1865 AGFG1 1893 SLC35F5
1922 C3orf35 1951
GPR17//L0C10029 1866 CHRNG 1894 CCDC93
1923 SNRK 1952
1428//LIMS2 EIF4E2 1895 CLASP1
1924 ZNF197 1953 0
IMP4 1867
n.)
TRPM8 1896 SAP130
1925 GNAI2 1954 o


FAM123C 1868
un
LRRFIP1 1897 YSK4
1926 ALAS1 1955 1¨

ACVR2A 1869
1-
--4
GAL3ST2 1898 GTDC1
1927 PRKCD 1956 tµ.)
MBD5 1870
o
.6.
TMEM18 1899 ORC4L
1928 CACNA1D 1957
LYPD6B 1871
LAPTM4A 1900 NR4A2//F1146875
1929 PXK 1958
SLC4A10 1872
SF3B14 1901 DPP4
1930 PTPRG 1959
UBR3 1873
TP5313 1902 GALNT3
1931 ATXN7 1960
HAT1 1874
UNQ2999 1903 SCN7A
1932 SLC35A5 1961
ITGA6 1875
GPR113//SELI 1904 FRZB
1933 SLC15A2 1962
ZAK 1876
MPV17 1905 STK17B
1934 CCDC48 1963 P
OSBPL6 1877
PPM1G 1906 CLK1//PPIL3
1935 DNAJC13 1964 .
"
PLEKHA3 1878
.
NLRC4 1907 MPP4
1936 CLDN18 1965 .
,
ZC3H15 1879
N,
u,
CDC42EP3 1908 IN080D
1937 GYG1 1966 tv "
COL3A1 1880
,
,
HNRPLL 1909 KLF7
1938 SELT 1967 1
GLS 1881
.
u,
,
COX7A2L 1910 FAM119A
1939 MED12L 1968 ,
OBFC2A 1882
,
KCNG3 1911 NGEF
1940 RAP2B 1969
COQ10B 1883
CALM2//C2orf61 1912 ARL4C
1941 MYNN 1970
MARS2 1884
BCL11A 1913 RAB17
1942 ABCF3 1971
CFLAR 1885
XPO1 1914 HDLBP
1943 VPS8 1972
NOP58 1886
NAT8B 1915 LRRN1
1944 HRG 1973
FAM117B 1887
DUSP11 1916 SETD5
1945 EIF4A2//SNORA4 1974 1-d
CYP20A1 1888
n
MOGS 1917 IRAK2
1946 LPP 1975 1-3
FASTKD2 1889
5;
SNRNP200 1918 C3orf42
1947 CCDC50 1976 t.)
PIKFYVE 1890
SEMA4C 1919 TSEN2
1948 L0C152217 1977 1¨

un
C2orf62 1891
-,-:--,
MITD1 1920 NR2C2//MRPS25
1949 TADA3L 1978 un
SLC11A1 1892
o
IL1A 1921 UBE2E1
1950 SEC13 1979 .6.
c,.)

TIMP4 1980 FOXL2 2007 RBM46
2034 CARD6 2062
METTL6 1981 MECOM 2008 GRIA2
2035 MCCC2 2063
DAZL//DAZ4//DAZ 1982 PLD1 2009 C4orf39
2036 TNP01 2064 0
3//DAZ2
tµ.)
o
GNB4 2010 KLHL2
2037 PDE8B 2065 1¨

SATB1//TBC1D5 1983
un
MRPL47 2011 TLL1
2038 PAPD4 2066 1¨

SCN10A 1984
1-
--4
KLHL6 2012 F11
2039 THBS4 2067 tµ.)
SEC22C 1985
o
.6.
THPO 2013 SLBP
2040 FAM151B 2068
ZDHHC3 1986
ETV5 2014 HAUS3//POLN
2041 RASGRF2 2069
ZDHHC3 1987
BCL6//LOC100131 2015 PPARGC1A
2042 SNX2 2070
SLC6A20 1988 635 TLR10
2043 LMNB1//PCIF1 2071
UQCRC1 1989 ATP13A5 2016
C4orf34
2044 MEGF10 2072
PRKAR2A 1990 TMEM44 2017
TXK
2045 LEAP2 2073
IMPDH2 1991 KIAA1530 2018
RPL21P44
TCF7 2074 P
CCDC71 1992 TACC3 2019
KDR
2046 KDM3B 2075 .
"
UBA7 1993 CNO 2020
w
RCHY1
2047 CXXC5 2076 .
N,
CAMKV 1994 BST1 2021
u,
CNOT6L
2048 SLC4A9 2077 tv "
WDR82 1995 KLF3 2022
'7-1 .
,
,
PLAC8
2049 ANKHD1- 2078 1
LMOD3 1996 TMEM33//DCAF4L 2023
.
u,
EIF4EBP3//ANKHD
,
1 HPSE
2050 ,
,
FOXP1 1997
1//EIF4EBP3
KIT 2024 GPRIN3
2051 KIAA0141 2079
MORC1 1998
ENAM 2025 PPA2
2052 GRPEL2 2080
ATG3 1999
FAM47E//STBD1 2026 COL25A1
2053 MFAP3 2081
GSK3B//LOC10012 2000
9275 ENOPH1 2027 C4orf3
2054 GABRA6 2082
HCLS1 2001 PDLIM5 2028 QRFPR
2055 GABRA1 2083
KPNA1 2002 CCDC109B//HIGD 2029 MFSD8
2056 DOCK2 2084 1-d
n
1A//CCDC13
1-3
PTPLB 2003 MAP9
2057 RANBP17//USP12 2085
EGF 2030
5;
C3orf22 2004 PDGFC
2058t.)
PCDH10 2031
ERGIC1 2086
RPN1 2005 TKTL2
2059 ATP6V0E1//SNORA 1¨
RAB33B 2032
un
KIAA1257//ACAD9 2006 ACSL1
2060 74B -c-:--,
un
//LOC100132731 TMEM184C 2033
ZNF346 2087
o
SUB1//TMEM183A 2061
.6.
c,.)

NSD1 2088 ADAM19 2115 ZNF451
2143 YIPF3 2171
CLPTM1L 2089 SLIT3 2116 SOBP
2144 MRPL14 2172
UGT3A1 2090 RNF44 2117 C6orf182
2145 PLA2G7 2173 0
GDNF 2091 DOK3 2118 KIAA1919
2146 PKHD1 2174 o

vi
TTC33 2092 MGAT4B//SQSTM1 2119 RWDD1
2147 IL17F 2175 1-
1-
--4
hCG 2039148 C5orf45//SQSTM1 2120 KPNA5
2148 HTR1B 2176 t,.)
o
.6.
MOCS2 2093 RASGEF1C 2121 TPD52L1
2149 GABRR2 2177
SLC38A9 2094 MGAT1 2122 ARG1
2150 UBE231 2178
CCDC125 2095 IRF4 2123 RAB32
2151 BACH2 2179
ANKRA2 2096 HIVEP1 2124 ARID1B
2152 MCM9 2180
HAPLN1 2097 E2F3 2125 SLC22A3
2153 VNN1 2181
CCNH 2098 HIST1H4I 2126 SERPINB1
2154 IL2ORA 2182
TMEM161B 2099 HIST1H2BM 2127 C6orf146
2155 F1127255 2183 P
MBLAC2 2100 MOG 2128 GCM2
2156 T 2184 .
N,
w
MCTP1 2101 ZNRD1//NCRNA00 2129 ATXN1
2157 RPS6KA2 2185 .
N,
171
TICAM2//TMED7// 2102 DCDC2//KAAG1 2158
HGC6.3 2186 , "
TMED7-TICAM2 TRIM15 2130
00 .
,
,
HIST1H3I
2159 UNC84A//C7orf20 2187 .
KIF3A 2103 HCG27 2131
.
u,
,
HIST1H4L
2160 SDK1 2188 ,
C5orf15 2104 BAT2//SNORA38 2132
GABBR1
2161 ZDHHC4 2189
SKP1 2105 CYP21A2 2133
RNA243
C7orf26 2190
CXCL14 2106 ITPR3 2134
DDAH2
2162 GLCCI1//tcag7.90 2191
KLHL3 2107 MAPK14 2135
3
CLIC1
2163
CD14 2108 MAPK13 2136
GPNMB 2192
NEU1
2164
YIPF5 2109 PNPLA1 2137
CCDC126 2193
RXRB
2165 1-d
LARS 2110 SFRS3 2138
WIPF3//ZNRF2//L 2194 n
VPS52
2166 0C441208 1-3
DCTN4 2111 CDKN1A 2139

TCP11
2167 GPR141 2195 t.)
CCDC69 2112 FOXP4 2140
CLPS
2168 STARD3NL 2196 1¨

ATOX1 2113 CUL9 2141
vi
PGC
2169 POU6F2 2197 -a-,
vi
TIMD4 2114 RUNX2 2142
ZNF318
2170 CDC2L5 2198 o
.6.
c,.)

ZMIZ2 2199 INHBA 2224 F1146365 N/A
TGFBR1 2278
UPP1 2200 CAMK2B 2225 SNTG1
2251 C9orf6//IKBKAP 2279
ZNF273 2201 NPC1L1 2226 TRIM55
2252 1MAGE5303689 N/A 0
KCTD7//RABGEF1 2202 DDC//L0C100129 2227 C8orf45
2253 ATP6V1G1 2280 o
1-
427
vi
RABGEF1//tcag7.9 2203 PREX2
2254 TLR4 2281 1-
67//tcag7.951//KC NSUN5//NSUN5B// 2228
1-
--4
TD7//L0C1002933 NSUN5C PLEKHF2
2255 SET 2282 t,.)
o
.6.
33 CLDN3 2229 BAALCHFL310489 2256
MRPL41 2283
CCDC132 2204 C7orf23//DMTF1 2230 11C35
2257 C9orf68 2284
PVRIG//PILRB//ST 2205 SRI 2231 MTBP
2258 HAUS6//SCARNA8 2285
AG3 BET1 2232 ZHX2
2259 KLHL9 2286
PILRB//PVRIG//ST 2206
AG3 MCM7 2233 RNF139
2260 C9orf82 2287
C7orf51 2207 GATS 2234 TG
2261 NDUFB6//DFFB 2288
GNB2 2208 ATXN7L1//RINT1// 2235 DENND3//C8orf60
2262 SIT1 2289 P
LRRC17 2209 EFCAB10
.
TNFRSF1OD
2263 FAM108B1 2290 "
KIAA1549 2236
'
w
LRRN3 2210
0
TRIM35
2264 TRPM6 2291 .
5LC37A3 2237
"
u,
CFTR 2211
tv
GSR
2265 FRMD3 2292 "
SMARCD3 2238
,
LSM8 2212
,
WHSC1L1
2266 5LC28A3 2293 1
MLL3//BAGE2 2239

u,
LUC7L2 2213
'
PCMTD1//PXDNL 2267
BICD2 2294 ,
CLN8 2240
-J
MGAM//LOC10012 2214 NCOA2
2268 C9orf84 2295
4692 MSRA 2241
GIMAP7 2215 PIWIL2 2242 TRAM1//L0C28619
2269 AKNA 2296
0
INSIG1 2216 NEFM//L0C100129 2243 RUNX1T1
2270 MEGF9 2297
RBM33 2217 717
C5 2298
EXT1
2271
EPHX2 2244
ICA1 2218
GOLGA1//SCAI 2299
DDEF1IT1
2272
LEPROTL1 22451-d
FAM126A 2219
SH2D3C 2300
CDC37L1
2273 n
MAK16//C8orf41 2246
1-3
HIBADH 2220
FAM102A 2301
UBE2R2
2274 5;
AP3M2 2247
TRIL 2221
F1110232 N/A t.)
UBAP1//KIF24
2275
FNTA 2248
SCRN1 2222
ASB6 2302 1¨

vi
GALT
2276
5GK196 2249'a
ELMO1 2223
BAT2L 2303 vi
RGP1//GBA2
2277
UBE2V2 2250
EDF1 2304 =
.6.
c,.)

FBXW5 2305 ATAD1 2332 PPFIA1
2360 CREBZF 2389
C10orf18 2306 ANKRD22 2333 IL18BP//NUMA1
2361 PICALM 2390
FBX018 2307 F1134077 N/A C11orf30
2362 SLC36A4 2391 0
tµ.)
GATA3 2308 COX15 2334 C11orf82
2363 CCDC82 2392 o


un
CUGBP2 2309 ERLIN1 2335 TMEM126B
2364 KIAA1826 2393 1-
1-
--4
VIM 2310 ACTR1A 2336 C11orf73
2365 MPZL3 2394 tµ.)
o
.6.
STAM 2311 ABLIM1 2337 PIWIL4
2366 MPZL2 2395
WAC 2312 RAB11FIP2 2338 L0C100132686
2367 H2AFX 2396
BAMBI 2313 C10orf84 2339 PAFAH1B2
2368 SIAE 2397
ZNF487//L0C4399 2314 PRDX3 2340 UBE4A
2369 ZBTB44 2398
11 C10orf119 2341 TRAPPC4
2370 HSN2 2399
ALOX5 2315
NSMCE4A 2342 SC5DL
2371 ADIPOR2 2400
WDFY4 2316
TALD01//INTS8 2343 VWA5A//0R10D1P 2372
NCAPD2//SCARNA 2401 P
SRGN 2317
TNNT3 2344 STT3A
2373 10//FADS1 .
"
CCDC109A 2318
PTPN6 2402 .
FXC1 2345 VPS26B
2374 .
FAM149B1//FAM14 2319
CLEC4D 2403 N,
u,
9B2 PDE3B 2346 TRIM21
2375tv
tv
"
.
CDKN1B
2404 ,
MINPP1 2320 DNAJC24 2347 ZBED5
2376 .
'
1
GOLT1B
2405 .
u,
,
PTEN//PTENP1 2321 PTPRJHOR4B1 2348 SAAL1
2377 ,
,
FAR2
2406
ENTPD1//C10orf13 2322 C11orf31 2349 FANCF
2378
1
FGD4 2407
TMEM109 2350 LIN7C
2379
ABCC2 2323
TMEM106C 2408
CD6 2351 PHF21A
2380
SFXN2 2324
TMBIM6 2409
CD5 2352 CUGBP1
2381
SHOC2 2325
C12orf62 2410
TMEM138 2353 OSBP
2382
ACSL5 2326
PRR13//PCBP2 2411
POLR2G 2354 CYBASC3
2383 1-d
BCCIP//DHX32 2327
DGKA 2412 n
TMEM179B 2355 TUT1
2384 1-3
FAM188A 2328
COQ10A 2413 5;
NAT11 2356 SLC25A45
2385 t.)
CUBN 2329
TSPAN31 2414
OTUB1 2357 LTBP3
2386 1¨

SVIL//hCG 17834 2330
CDK4/MARCH9/C3 2415 un
-,-:--,
94 RBM14//RBM4 2358 EIF1AD
2387 HC4 un
o
FAM13C//PHYHIPL 2331 AIP 2359 GAB2
2388 LEMD3 2416
.6.
c,.)

I RAK3 2417 PPTC7 2445 CCDC92
2473 ZMYM5//ZMYM2 2501
TMTC3 2418 DDX54//CCDC42B 2446 NCOR2
2474 ZDHHC20//L0C72 2502
ACTR6 2419 SLC24A6 2447 DHX37
2475 8099 0
PARP4
2503 t.)
TCTN1 2420 SDS 2448 DDX51
2476 o
1-,
MTMR6//L0C6464 2504
un
PXM P2//PGAM 5 2421 RBM19 2449 POLE
2477 82
1-,
DCP1 B 2422 MED13L 2450 GOLGA3
2478 HSPH1 2505 -4
t=.)
o
.6.
SLC2A3//SLC2A14 2423 C12orf49 2451 ZMYM2
2479 N4BP2L2//CG030 2506
C3AR1 2424 FBX021 2452 SPATA13//C1QTNF
2480 ELF1 2507
PLBD1 2425 WSB2 2453 9
LCP1 2508
N U PL1
2481
TM7SF3 2426 TAOK3 2454
KPNA3 2509
PAN 3//EEF1A1//C
2482
ASB8//PHB 2427 CIT 2455 HCHD2
C13orf1 2510
LMBR1L 2428 RAB35 2456 ALOX5AP
2483 DLEU2//DLEU2L 2511
FM N L3//PRPF4OB 2429 RPLPO 2457 EEF1DP3
2484 GUCY1B2 2512 P
AAAS 2430 PXN 2458 KL
2485 I NTS6 2513
N,
u,
L.
NFE2 2431 TRIAP1 2459 U FM1
2486 DACH1 2514 .
0
ND
U1
GPR84 2432 SFRS9 2460 NARG1L
2487 TBC1 D4 2515 tv
t.)
ND
0
CD63 2433 POPS 2461 ITM2B
2488 EDNRB 2516 cn
,
1
.
u,
,
SARNP//DNAJC14 2434 UNQ1887 2462 FNDC3A
2489 UGGT2 2517 ,
,
NACA 2435 C12orf43 2463 CDADC1
2490 GPR183 2518
CDK4//TSPAN 31 2436 ANAPC5 2464 ARL11
2491 LIG4 2519
TM BIM4//LOC1001 2437 KDM2B 2465 LMO7
2492 ANKRD10 2520
33322
MORN3 2466 DNAJC3
2493 RASA3 2521
1L22 2438
TM EM 120B//RHOF 2467 TM9SF2
2494 RNASE2//L0C6433 2522
LIN7A 2439
32
L0C338799 CLYBL
2495 Iv
HAL 2440
RPGRIP1 2523 n
DIABLO//B3GNT4 2468 PCCA
2496 1-3
APPL2 2441
IRF9 2524 5;
VPS33A 2469 ABHD13
2497 t,.)
GLTP 2442
TSSK4 2525
CLIP1 2470 LAMP1
2498
GIT2 2443
C14orf21 2526 un
PITPN M2 2471 TMC03
2499 'a
un
VPS29 2444
SCFD1 2527
EIF2B1 2472 UPF3A
2500 o
.6.
c,.)

FANCM 2528 NDRG2 2554 BCL11B
2583 SNX1 2610
ABHD12B 2529 DAD1//OR631 2555 AN KRD9
2584 LBXCOR1//PIAS1// 2611
PTGDR 2530 SLC7A8 2556 PPP1R13B
2585 CALM L4 0
tµ.)
FBX034//K1AA083 2531 1PO4 2557 AKT1
2586 NE01 2612 o
1

M PI
2613 un
TM9SF1 2558 BRF1
2587 1¨

C14orf101 2532
FBX022//FBX022 2614 1¨
--4
ADCY4 2559 TUBGCP5
2588 OS tµ.)
o
ACTR10 2533
.6.
RI PK3 2560 SNRPN *
2589 RCN2 2615
ARID4A 2534
EAPP 2561 APBA2
2590 FAH 2616
3 KAMP 2535
BAZ1A 2562 MTMR15//MTMR10 2591
IL16 2617
HIF1A 2536
NFKBIA 2563 RYR3
2592 ABHD2 2618
SYNE2 2537
SEC23A 2564 BAHD1
2593 SLCO3A1 2619
EXD2 2538
C14orf104 2565 CHP
2594 MCTP2 2620
5LC39A9 2539
C14orf138 2566 3M3D7-
2595 M EF2A//LYSM D4 2621 P
SFRS5 2540
.
5052 2567 PLA2G4B//3M3D7//
N I PA2//CYFIP1 2622 N,
PCNX 2541 PLA2G4B
w
w
NIN 2568 HAUS2
2596 HERC2//HERC2P2/ 2623 .
ND
SIPA1L1//SNORD5 2542
/HERC2P3//LOC44
6B//L0C145474//L PYGL 2569 C15orf63//SERF2
2597 0248 tv "
tv
,
0C283567 CNIH 2570
YLPM1 2543 B2M
2598 MTMR10//MTMR15 2624 1 .
u,
,
DHRS7 2571 TRI M69
2599 C15orf24 2625 ,
,
BATF 2544
WDR89 2572 PLDN
2600 SLC12A6 2626
FLVCR2//RPS24 2545
ACTN1 2573
GPR65 2546 SQRDL
2601 LPCAT4 2627
NUMB 2574 GALK2
2602 1N080 2628
TDP1 2547
C14orf43 2575 USP8
2603 01P5 2629
EVL 2548
ABCD4 2576 GLDN
2604 ZFP106 2630
ZN F839 2549
1-d
KIAA0317 2577 MAPK6
2605 CDAN1 2631 n
TDRD9 2550
1-3
NEK9 2578 LACTB
2606 SPG11//ISLR 2632
5;
IN F2 2551
ANGELI. 2579 RAB8B
2607 SPPL2A 2633 t.)
PLD4 2552
SPTLC2 2580 APH1B
2608 GNB5//L0C100129 2634 1¨

un
MTA1//L0C647310 2553
973 'a
USP3//LOC100130 2609
un
//L0C100128343 SERPINA6 2581
o
DICER1 2582 855
MY05A 2635 o
.6.
c,.)

ARPP19 2636 KCTD5//PR00461/ 2664 CMTM1//CKLF
2691 CLCN7 2719
RAB27A 2637 /PDPK1 TM EM208
2692 MRPS34 2720
CLUAP1 2665
CCPG1//PIGB//DY 2638 CTCF
2693 RN PS1 2721 0
X1C1 DNASE1 2666
tµ.)
o
BNIP2 2639 DNAJA3 2667
THAP11
2694 NLRC3 2722 1¨
un
CA12 2640 CP110 2668
NUTF2
2695 TRAP1//DNASE1 2723 1¨
1-
--4
tµ.)
FAM96A 2641 C16orf62 2669 EDC4
2696 ADCY9 2724 o
.6.
KIAA0101//CSN K1 2642 LYRM 1 2670 SLC7A6//SLC7A60
2697 COR07 2725S
G1
C16orf72 2726
M ETT L9 2671 PRMT7
2698
TLE3 2643
RRN3//L0C653390 2727
EEF2K 2672 SNTB2
2699
PARP6 2644
//L00730092//L0
POLR3E 2673 VPS4A
2700 C100131998
NPTN 2645
PLK1 2674
MAN2C1 2646 DDX19B//DDX19A 2701
XYLT1//LYRM2//Z 2728 PRKCB 2675 CHST4 2702 C3H11ADCU N 1D3//LYRM
1 2729
IMP3 2647
P
IL21R//L0C28388 2676 HP//HPR
2703 IGSF6//METTL9 2730
MTHFS 2648
.
8
N,
w
ST20//C15orf37 2649 SULT1A2//SULT1A 2677 PLCG2
2704 CDR2//RRN3//LOC 2731 .
1 KLHL36
2705 100131998//LOC6 "
u,
TMC3 2650
tv "
ATXN2L 2678 KIAA0182
2706 53390 tv
,
AP3B2 2651
COG7 2732
,
LAT II 2679 BAN P//RUN DC2C
2707 .
u,
,
C15orf40 2652
GGA2 2733 ,
KIF22 2680 TRAPPC2L
2708 ,
WDR73 2653
NTRK3 2654
NSMCE1 2734
MAZ 2681 SPG7
2709
GTF3C1
2735
CORO1A//L0C606 2682 CDK10
2710
DET1 2655 724
CCDC101//L0C388 2736
TM2D3 2656 ITGAL 2683 TCF25
2711 242
WDR90 2657 SRCAP//SNORA30 2684 AFG3L1
2712 C16orf54 2737
RHOT2//FBXL16 2658 ZN F646//ZN F668 2685 LUC7L
2713 KCTD13 2738 1-d
n
TM EM204 2659 C16orf67 2686 AXIN 1
2714 SEPT1 2739 1-3
CRAMP1L//HN1L 2660 TM EM188 2687 3M3D8
2715 ZN F764//ZN F747 2740 5;
t.)
LM F1
2716 C16orf58//LOC100 2741
MAPK8IP3 2661 LPCAT2 2688
128371 1¨
UNKL
2717 un
TBL3 2662 CETP 2689
ITFG1 2742 'a
un
TSC2 2663 CKLF 2690 UNKL
2718 ABCC11//LON P2 2743 o
.6.
c,.)

N U DT21 2744 SGSM2 2772 KPN B1
2799 TMC8 2826
BBS2//0GFOD1 2745 ARRB2 2773 CDK5RAP3
2800 ENGASE 2827
CSNK2A2 2746 L0C100130950 2774 ATP5G1
2801 RPTOR 2828 0
r..)
GOT2 2747 DNAH2 2775 UBE2Z
2802 GPS1 2829 o


un
FAM96B 2748 PIGL 2776 XYLT2//LOC10013
2803 FN3KRP 2830 1¨

-4
FHOD1//SLC9A5 2749 TRPV2 2777 0580
TBCD 2831 r..)
NOG
2804 o
.6.
ATP6V0D1//LOC10 2750 MPRIP 2778
GEMIN4 2832
0132855 DGKE
2805
DRG2 2779
GLOD4 2833
GFOD2 2751 AKAP1
2806
ALKBH5//Fll 1377 2780
SLC43A2 2834
SLC12A4 2752 3 TM EM49//CLTC//M
2807
IR21
PRPF8 2835
DPEP3 2753 SMCR7 2781
C LTC
2808 SMG6//C17orf6 2836
DPEP2 2754 WSB1 2782
CA4
2809 METT10D//L0C284 2837
CHTF8//HAS3 2755 TAOK1 2783
C17orf64
2810 009
P
COG8//PDF 2756 CPD 2784
SHPK 2838
DCAF7
2811 .
N,
TERF2 2757 SUZ12P 2785
TAXI BP3 2839 u,
L.
PITPNC1
2812 .
u,
AARS 2758 RN F135 2786
P2RX5 2840 N,
u,
NOL11//SNORA38 2813
ST3GAL2 2759 ZN F830 2787 B
MYBBP1A//SPNS2 2841 tv
-1.
.
,
0,
,
VAC14//LOC10013 2760 TAF15 2788 MAP2K6
2814 PELP1 2842
u,
,
0894,
,
GGNBP2 2789 COG1
2815 PFN1 2843
AP1G1 2761
LASP1 2790 CD300A
2816 ZN F232 2844
WDR59 2762
PSM D3 2791 TM EM104
2817 DHX33 2845
CTRB2//CTRB1 2763
CDC6 2792 MRPS7
2818 DERL2 2846
TAF1C//ADAD2 2764
NBR2 2793 KIAA0195
2819 NLRP1//L0C72839 2847
FBX031 2765
TM UB2 2794 TSEN 54
2820 2
ZCCHC14 2766
ASGR2 2848 1-d
MGC57346//C17or 2795 LLGL2
2821 n
FAM 38A 2767 f69 L0C100134934//C
2822 N EU RL4//GPS2//D 2849
4S234E
1-3
5;
CEN PBD1 2768 NSF//L0C728806 2796 DK3
t,.)
ZBTB4
2850
TIMM22 2769 GOSR2 2797 MFSD11
2823 1¨

TP53
2851 un
RPA1 2770 NPEPPS//TBC1D3F 2798 SEPT9
2824 'a
un
//L0C440434
__________________________________________________________________________ VAM
P2 2852
DPH1//0VCA2 2771 TN RC6C
2825 o
.6.
c,.)

PIK3R5 2853 UBTF 2879 EXOC7
2904 PSTPI P2 2931
ELAC2 2854 GPATCH8 2880 SFRS2
2905 C18orf32 2932
NCOR1//C200rf19 2855 MAP3K14//LOC100 2881 TMC6//LOC100131
2906 MBD2//SNORA37 2933 0
1//L0C100131704 133991 096
tµ.)
o
PIGN
2934 1¨

ZN F287 2856 OSBPL7 2882 USP36
2907 un
TMX3
2935 1¨

TOM1L2//L0C2463 2857 SLC35B1 2883 CD7
2908 1-
--4
15
PQLC1 2936 tµ.)
TOB1 2884 RAB31
2909 o
.6.
GRAP//SNORD3B- 2858
GZMM 2937
1//SNORD3B- COX11//TOM1L1 2885 VAPA
2910
2//L0C400581 VEZ F1 2886 SEH1L
2911 ARID3A 2938
ALDOC 2859
CIRBP 2939
SFRS1//F1144342 2887 HQ0644/PR00644 2912
SDF2 2860
DAZAP1 2940
SEPT4 2888 RN MT
2913
RAB34 2861
SPPL2B 2941
MED13//LOC1001 2889 RN F138
2914
PHF12 2862 29112
NFIC 2942
GALNT1
2915
N U FIP2 2863 LIM D2//MAP3K3 2890
VAV1 2943 P
ELP2
2916
OMG 2864 STRADA 2891
ARHGEF18//LOC10 2944 0
"
PIK3C3
2917 .
EVI2B 2865 FTS33 2892
0128573 .
N,
SLC14A2
2918 STXBP2//L0C5543 2945 u,
C17orf66//RSL24D 2866 CD79B 2893
63//L0C10013180
tv .
ME2
2919 CA
1 ICAM 2 2894
1 .
,
SYNRG//LOC1001 2867 SERPINB2//SERPI
2920 C19orf59 2946 .
u,
,
ERN 1 2895
,
NB10
31822
,
PLXDC1 2868 TEX2 2896 ZN F407
2921 ZN F317 2947
I LF3
2948
CACN B1 2869 LRRC37A3//LRRC3 2897 ZN F236
2922
7A2//LRRC37A//A
SMARCA4 2949
PGAP3 2870 NFATC1//LOC1001
2923
RL17P1//LRRC37A 27994
PRKCSH 2950
MED24 2871 4//L0C100294335
ENOSF1//TYMS
2924
NR1D1//THRA 2872 //L00644397
I ER2 2951
GNA13 2898 MYOM 1
2925 CCDC130 2952
CCR7 2873
1-d
WIPI1//ARSG 2899 AFG3L2
2926 DCAF15 2953 n
STAT5B//STAT5A 2874
1-3
FAM 20A 2900 ABHD3
2927 IL27RA 2954 5;
FAM 134C 2875
t.)
NAT9 2901 OSBPL1A
2928 KLF2 2955
VAT1 2876


GGA3 2902 CDH2
2929 SIN3B 2956 un
'a
DUSP3 2877
un
H3F3B//H3F3C 2903 DSC1
2930 DDA1 2957
o
C17orf65//ASB16 2878
.6.
c,.)

GTPBP3 2958 VASP 2984 PTPRS
3009 ZNF529 3036
FAM129C 2959 GRWD1 2985 SAFB2//SAFB
3010 ZNF461 3037
FCH01 2960 FLT3LG 2986 RANBP3
3011 ZNF607 3038 0
tµ.)
ARRDC2 2961 ZNF175 2987 GTF2F1//L0C1001
3012 YIF1B 3039 o


un
30856
1F130 2962 NCRNA00085
PRR13 3040 1¨

XAB2
3013 1-
--4
C19orf60 2963 PPP2R1A
CEACAM4 3041 tµ.)
ELAVL1
3014 o
4=.
CRTC1//MAML2 2964 ZNF808//ZNF578/ 2988
PLAUR 3042
ADAMTS10
3015
RFXANK//MEF2B// 2965 /ZNF611
TRAPPC6A 3043
LENG8 2989 FBXL12
3016
L00729991
ERCC1//CD3EAP 3044
ZNF101 2966 FCAR 2990 DNMT1
3017
RTN2
3045
ZNF738 2967 RPL28 2991 TYK2
3018
SYMPK
3046
ZNF257//ZNF492/ 2968 U2AF2 2992 KEAP1
3019
/ZNF99//ZNF98//L
PGLYRP1
3047
L0C100288114//M KRI1
3020
00646864
NOSIP 3048
GC9913
P
C19orf2 2969 TMEM205//hCG 2 3021
.
ZFP28 2993 9977
PNKP 3049 "
u,
K1AA0355//F11213 2970
w
ZNF460 2994 ZNF563
3022 NKG7 3050 u,
69
U 1"
USF2 2971 ZNF549 2995 MAN2B1//MORG1 3023
FPR1 3051 tv
tv
"
.
TMEM147 2972 ZNF211 2996 C19orf56
3024 ZNF28 3052 . .
u,
LIN37//PSENEN 2973 ZNF587//ZNF417 2997 DHPS
3025 OSCAR 3053 ,
,
,
C19orf55 2974 ZNF274 2998 TNP02//SNORD41 3026
MBOAT7 3054
TBCB//POLR2I 2975 ZNF544 2999 LPHN1
3027 LILRA5 3055
ZNF382 2976 ZNF8 3000 NDUFB7
3028 LILRA4 3056
ZNF568 2977 TRIM28 3001 AKAP8
3029 ZNF550//ZNF549 3057
ZNF420 2978 C19orf6 3002 AKAP8L
3030 ZNF416 3058
ZNF383 2979 C19orf34 3003 CHERP//C19orf44/
3031 ZNF256 3059 1-d
n
1-3
CCDC97 2980 GNG7 3004 /CALR3
ZNF329 3060
INSL3//JAK3
3032 5;
ZNF574 2981 AES 3005
FAM110A 3061 t.)
IL12RB1
3033
CD177 2982 EEF2//SNORD37 3006
ITPA 3062 1¨

un
UPK1A
3034 'a
ZNF230//ZNF222 2983 PLIN5//LRG1 3007
TYROBP
3035 CDC25B 3063 un
PLIN3 3008
CDS2 3064 o
.6.
c,.)

CRLS1 3065 CSTF1 3092 B4GALT5
3119 KCNE1 3147
CSRP2BP 3066 CASS4 3093 DPM1
3120 PRDM15 3148
SEC23B 3067 GNAS 3094 ZFP64
3121 C2CD2 3149 0
tµ.)
SLC24A3 3068 C200rf177 3095 ZNF217
3122 WDR4 3150 o


un
HCK 3069 CDH26 3096 CTSZ
3123 U2AF1 3151 1-
1-
--4
ASXL1 3070 C200rf197 3097 SYCP2
3124 CSTB 3152 tµ.)
o
.6.
ACSS2 3071 L0C284757 PSMA7
3125 UBE2G2//SUM03 3153
C200rf4 3072 ARFGAP1 3098 DIDO1
3126 PTTG1IP 3154
TGIF2 3073 PRPF6 3099 YTHDF1
3127 POFUT2 3155
C200rf24//SLA2 3074 NSFL1C 3100 CHODL
3128 MCM3AP 3156
RPN2//EEF1A2 3075 SIRPD 3101 BACH1
3129 IL17RA//CECR7 3157
CTNNBL1 3076 SIRPG//SIRPA 3102 C21orf41//BACH1
3130 C22orf37 3158
ACTR5 3077 RNF24 3103 IL1ORB
3131 LZTR1 3159 P
PPP1R16B 3078 RASSF2 3104 IFNAR1
3132 PPIL2//YPEL1 3160 .
"
u,
DHX35 3079 TMX4 3105 IFNGR2
3133 CYTSA 3161 .
u,
N,
u,
PLCG1 3080 JAG1 3106 SON
3134 SNRPD3//C22orf1 3162 tv
tv
"
MYBL2 3081 C200rf74 3107 MORC3//DOPEY2 3135
,
1
NF2
3163 .
u,
,
SYS1//SYS1- 3082 C200rf3 3108 DYRK1A
3136 ,
,
DBNDD2//DBNDD
LIMK2 3164
C20orf112 3109 KCN315
3137
2
SLC5A1 3165
DNTTIP1 3083 CDK5RAP1 3110 ETS2
3138
MCM5
3166
CTSA 3084 AHCY 3111 RRP1B
3139
NCF4
3167
MMP9//LOC10012 3085 GGT7 3112 PFKL
3140
GGA1
3168
8028 EDEM2 3113 TRPM2
3141
DDX27 3086
SH3BP1//PDXP 3169
RBM39//L0C64316 3114 ADARB1
3142 1-d
SLC9A8 3087 7
POLR2F//LOC1001 3170 n
SAMSN1//L0C388 3143
31530 5;
RNF114 3088 BLCAP 3115 813
APOBEC3A//APOB 3171 t.)
PTPN1 3089 SERINC3//TTPAL 3116 N6AMT1
3144 EC3B 1¨

TSHZ2 3090 ZNF335 3117 SYN31
3145 APOBEC3D 3172 un
-c-:--,
un
PFDN4 3091 ELMO2 3118 TMEM5OB
3146 ATF4 3173
o
.6.
c,.)

CACNA1I 3174 MKL1//KIAA1659 3200 UPRT
3228 MA0B//NAT13 3256
ZC3H7B 3175 RANGAP1 3201 CXorf26
3229 ZN F41 3257
CCDC134 3176 TCF20 3202 ATP7A
3230 OTUD5 3258 0
TSPO 3177 LDOC1L 3203 DIAPH2
3231 KCN D1 3259 o

vi
N U P50 3178 UNQ6126 3204 CSTF2//RAD21
3232 ZMYM3 3260 1-
1-
--4
TBC1D22A//LOC10 3179 TUBGCP6 3205 ARMCX3
3233 MAGT1 3261 t,.)
o
0289878
.6.
SBF1//SBF1P1 3206 ARMCX5
3234 BRWD3 3262
RP3-402G11.5 3180
MSL3 3207 GPRASP1
3235 TRMT2B 3263
SAPS2 3181
MOSPD2 3208 TM EM31
3236 GLA 3264
NCAPH2 3182
BMX//HNRPDL 3209 TBC1D8B
3237 MORF4L2 3265
BID 3183
PDHA1 3210 MID2
3238 PSM D10 3266
SLC25A1 3184
YY2 3211 DOCK11
3239 ACSL4 3267
KLHL22//KRT18 3185
PDK3 3212 LON RF3
3240 LAMP2 3268 P
PI4KA//PI4KAP1// 3186
PI4KAP2//LOC100 GK//GK3P//FTL//L 3213 UBE2A
3241 CU L4B 3269 .
"
293141 00652904 SH2D1A
3242 ODZ1 3270 w
N,
MAPK1 3187 CXorf59 3214
u,
OCRL
3243 ELF4 3271 tv "
tv
.
ZN F70 3188 ATP6AP2 3215
00 ,
,
SLC25A14
3244 RAP2C 3272 .
TPST2 3189 USP9X//USP9Y 3216
.
u,
,
HPRT1
3245 FAM127B//FAM127 3273 ,
_.]
SF3A1//CCDC157 3190 RP2 3217
C//FAM 127A
CD4OLG
3246
PES1 3191 USP11 3218
TM EM185A 3274
AFF2
3247
PIK3IP1 3192 RBM3 3219
ARD1A 3275
SSR4//IDH3G
3248
PATZ1 3193 FTS31 3220
IRAK1 3276
FAM 50A
3249
C22orf30 3194 WAS 3221
DNASE1L1//RPL10 3277
DKC1//SNORA36A 3250
IL2RB 3195 PLP2 3222 //SNORA56
SH3KBP1 3278
1-d
CSNK1E//L0C4009 3196 TSPYL2//GPR173 3223 ARSD
3251 M itochond ria I N/A n
1-3
27 MAGED2 3224 KALI.
3252 M itochond ria I N/A 5;
UNC84B 3197
t.)
U BQLN 2 3225 CTPS2
3253 CCN L2 3279
CBX7//L0C100128 3198


vi
NLGN3 3226 RPS6KA3
3254 IN PP5B 3280
400
'a
vi
RPS19BP1 3199 ACRC 3227 BCOR
3255 TLR5 3281
o
.6.
c,.)

ADRB3//GOT1L1 3282 NOC2L//SAMD11// 3283 LOC401010
SHFM1 3284
2fiSNORD115-39//SNORD116-
14//SNORD116-20//SNORD115-
0
HUGO Gene Nomenclature Committee
8fiSNORD115-3//SNORD115- n.)
o
38//SNORD115-41//SNORD115-
un
Synonymous with 22//SNORD115-44//SNORD116-
-4
SPNS1fiN PI PL2HL0C728741HL0C730153/ 1fiSNORD115-17//SNORD115-
n.)
o
IN PIPL3HSPIN1//L0C728888//L0C100289 18//SNORD115-19//SNORD115-
.6.
169HLOC728734//LOC729602MOC10028 20//SNORD116@
8442M0C100288332
* Synonymous with
SNURFHIPWHSNORD116-16//SNORD116-
18//SNOR 0116-21//SNOR 0116-
22//SNORD116-17//SNORD116-
19/PAR5HPAR-SNHSNORD116-
P
2fiSNORD116-25//SNORD116-

i.,
26//SNORD107//SNORD115-
'
12//SNORD115-5//SNORD115-
"
6fiSNORD115-9//SNORD116-
11//SNORD116-12//SNORD116-
1
i
.
u,
'
13//SNORD116-28//SNORD116-
,
...]
4fiSNORD64/PARMSNORD109AHSNORD
109BHSN0RD116-6//SN0RD116-
3fiSNORD116-9//SNOR 0115-
13//SNORD115-1//SNORD115-
14//SNOR 0115-15//SNOR 0115-
21//SNORD115-10//SNORD115-
7fiSNORD115-16//SNOR 0115-
40//SNORD115-42//SNORD115-
IV
n
11//SNORD115-29//SNORD115-
1-3
34//SNORD115-36//SNORD115-
5;
t.)
4fiSNORD115-43//HB11-52-24//SNORD116-
1¨,
5fiSNORD116-7//SNORD115-
un
-a 5
26//SNORD115-30//SNORD116-
un
o
15//SNORD116-8//SNORD115-
.6.
c,.)

Representative Drawing

Sorry, the representative drawing for patent document number 2930925 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2015-02-06
(87) PCT Publication Date 2015-08-13
(85) National Entry 2016-05-17
Dead Application 2021-08-31

Abandonment History

Abandonment Date Reason Reinstatement Date
2020-08-31 FAILURE TO REQUEST EXAMINATION
2020-08-31 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2016-05-17
Maintenance Fee - Application - New Act 2 2017-02-06 $100.00 2016-12-08
Maintenance Fee - Application - New Act 3 2018-02-06 $100.00 2017-12-08
Maintenance Fee - Application - New Act 4 2019-02-06 $100.00 2018-12-07
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
IMMUNEXPRESS PTY LTD
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2016-05-17 1 57
Claims 2016-05-17 42 2,113
Drawings 2016-05-17 37 937
Description 2016-05-17 229 11,921
Cover Page 2016-06-08 1 34
International Search Report 2016-05-17 7 265
National Entry Request 2016-05-17 2 59

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :